EP2340042B1 - Methods and compositions for the treatment of cancer - Google Patents

Methods and compositions for the treatment of cancer Download PDF

Info

Publication number
EP2340042B1
EP2340042B1 EP09783138.2A EP09783138A EP2340042B1 EP 2340042 B1 EP2340042 B1 EP 2340042B1 EP 09783138 A EP09783138 A EP 09783138A EP 2340042 B1 EP2340042 B1 EP 2340042B1
Authority
EP
European Patent Office
Prior art keywords
cells
chok
acid ceramidase
inhibitors
cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP09783138.2A
Other languages
German (de)
English (en)
French (fr)
Other versions
EP2340042A2 (en
Inventor
Ana RAMÍREZ DE MOLINA
Lourdes GARCÍA OROZ
Juan Carlos LACAL SANJUÁN
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Traslational Cancer Drugs Pharma SL
Original Assignee
Traslational Cancer Drugs Pharma SL
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Traslational Cancer Drugs Pharma SL filed Critical Traslational Cancer Drugs Pharma SL
Priority to PL09783138T priority Critical patent/PL2340042T3/pl
Priority to EP09783138.2A priority patent/EP2340042B1/en
Publication of EP2340042A2 publication Critical patent/EP2340042A2/en
Application granted granted Critical
Publication of EP2340042B1 publication Critical patent/EP2340042B1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4425Pyridinium derivatives, e.g. pralidoxime, pyridostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/191Tumor necrosis factors [TNF], e.g. lymphotoxin [LT], i.e. TNF-beta
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/34Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57423Specifically defined cancers of lung
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/112Disease subtyping, staging or classification
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/914Hydrolases (3)
    • G01N2333/978Hydrolases (3) acting on carbon to nitrogen bonds other than peptide bonds (3.5)
    • G01N2333/98Hydrolases (3) acting on carbon to nitrogen bonds other than peptide bonds (3.5) acting on amide bonds in linear amides (3.5.1)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/04Screening involving studying the effect of compounds C directly on molecule A (e.g. C are potential ligands for a receptor A, or potential substrates for an enzyme A)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/10Screening for compounds of potential therapeutic value involving cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • the invention relates to the field of therapeutics and, more in particular, to the field of cancer therapeutics using compositions containing several therapeutic compounds showing improved activity with respect to the compounds used individually.
  • Choline kinase is the first enzyme of the Kennedy pathway or the phospatidylcholine (PC) synthesis pathway. It acts by phosphorylating choline to phosphorylcholine (PCho) using adenosine 5'-triphosphate (ATP) as a phosphate group donor.
  • Ras genes form a family of the so-called oncogenes which have been widely studied since they are activated in 25-30% of all human tumors and in several of them in 90%. Ras proteins have an important role in the transmission of intracellular signals due to their involvement in the regulation of cell proliferation, terminal differentiation and senescence.
  • Hemicholinium-3 (HC-3) as a relatively potent and selective blocking agent ( Cuadrado A., et al., 1993, Oncogene 8: 2959-2968 , Jiménez B., et al., 1995, J. Cell Biochem. 57:141-149 ; Hernández-Alcoceba, R. et al., 1997, Oncogene, 15:2289-2301 ).
  • This choline homologue with a biphenyl structure has been used for designing new antitumor drugs.
  • HC-3 is a potent respiratory paralyzing agent, it is not a good candidate for its use in clinical practice.
  • Some derivatives having improved inhibitory activity of the ChoK and reduced toxic effects have been synthesized based on the structure of HC-3 by introducing structural modifications in this compound.
  • Bisquaternized symmetric compounds derived from pyridinium have also been found to inhibit PCho production in whole cells ( WO98/05644 ). However, these derivatives have high toxicity levels limiting their extended therapeutic application.
  • Drug resistance is a fundamental problem that limits the effectiveness of many chemotherapies currently used in cancer treatment.
  • Drug resistance can occur due to a variety of mechanisms, such as increased drug inactivation, decreased drug accumulation, drug efflux from cancer cells, enhanced repair of chemotherapy-induced damage, activation of pro-survival pathways and inactivation of cell death pathways ( Hersey P. et al., 2008, Adv Exp Med Biol. 2008;615:105-26 ). Drug resistance can be inherent to the tumour cells before the initiation of an antitumor treatment. In addition, specific drug resistance mechanisms can be activated after exposure of tumour cells to a particular treatment.
  • siRNA designed to this purpose recognizes both ChoK ⁇ and ChoK ⁇ species ( Mori et al., Cancer Res., 2007, 67:11284-11290 ) but only ChoK ⁇ and not ChoK ⁇ is a molecular target in oncology (International patent application WO2006108905 ), questioning the potential therapeutic use of this strategy.
  • compositions comprising a therapeutically effective amount of one or several agents inhibiting the expression and/or activity of the choline kinase alpha protein, together with any other active ingredient that does not inhibit the function of the choline kinase alpha protein have been described ( EP1889920A ).
  • the invention relates to a composition
  • a composition comprising, separately or together, a first component comprising one or more choline kinase inhibitors specific for the choline kinase alpha isoform and a second component selected from of one or more acid ceramidase inhibitors and one or more chemotherapeutic agents, wherein the chemotherapy agent is selected from the group consisting of a DNA-alkylating agent, an antimetabolite, a mitotic inhibitor, an anthracycline, a topoisomerase I inhibitor, a topoisomerase II inhibitor, cetuximab, gefitinib and imatinib.
  • the invention in a second aspect, relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a composition according to the invention with a pharmaceutically acceptable carrier or excipient and to the uses thereof in medicine and, in particular, for the treatment of cancer.
  • an inhibitor of acid ceramidase, a chemotherapeutic agent or a ligand for a death receptor to increase the sensitivity of a tumor cell to a choline kinase inhibitor.
  • a method for the identification of cancer patients resistant to the therapy with ChoK inhibitors comprising determining the levels of acid ceramidase in a sample from said patient wherein the patient is identified as being resistant to ChoK inhibitors when the acid ceramidase levels in said sample are higher than a reference sample.
  • a method for selecting a personalised therapy for a patient suffering from cancer comprising determining the levels of acid ceramidase in a sample from said patient wherein if the expression levels of acid ceramidase in said sample are higher than in the reference sample, the patient is candidate for being treated with a combination of a ChoK inhibitor and an acid ceramidase inhibitor.
  • example 1 of the present invention describes that different primary cultures derived from human Non Small Cell Lung Cancer tissues show a differential sensitivity towards MN58b, a known ChoK inhibitor ( Figure 1 ). Surprisingly, this resistance has been shown to be caused by an increase in the expression levels of acid ceramidase, as it has been shown in examples 2 and 3 of the present invention ( Figure 2 ).
  • the function of ChoK is to phosphorylate Cho to generate phophocholine (Pcho), a precursor of the major component of the plasma membrane, phosphatidylcholine (PC) ( Lacal JC., IDrugs.
  • SM sphingomyeline
  • example 4 of the present application shows that treatment of NSCLS-derived tumor cells with the acid ceramidase inhibitor NOE results in an increased sensitivity to ChoK inhibitors.
  • the combined use of the ChoK inhibitor MN58b or RSM-932A and the acid ceramidase inhibitor D-NMAPPD results in an improved antitumoral effect when compared with the use of the inhibitors in isolated manner or when compared with the combined used of a ChoK inhibitor and an inhibitor specific for alkaline ceramidase (see example 4).
  • the combined administration of acid ceramidase inhibitors and ChoK inhibitors would allow the use of lower dosages of the later compounds, thus leading to less undesired side effects.
  • the invention provides a composition (hereinafter the first composition of the invention) comprising, separately or together, a first component comprising one or more choline kinase inhibitors specific for the choline kinase alpha isoform and a second component comprising one or more acid ceramidase inhibitors.
  • composition refers to one or more compounds in various combinations according to alternative embodiments of this invention.
  • the composition comprises at least an acid ceramidase inhibitor and at least a ChoK inhibitor specific for the choline kinase alpha isoform.
  • Choline kinase refers to an enzyme which catalyses the phosphorylation of choline in the presence of ATP to produce phosphorylcholine (PCho) (EC 2.7.1.32).
  • exemplary choline kinases which can be inhibited according to the present invention include choline kinase alpha (as defined in UniProt under accession numbers P35790, 054804 and Q01134 for the human, mouse and rat proteins, respectively).
  • Acid ceramidase (N-acylsphingosine deacylase activity, EC 3.5.1.23), as used herein, is the lipid hydrolase responsible for the degradation of ceramide into sphingosine and free fatty acids within lysosomes.
  • the cells contains at least three types of ceramidases which are classified, according to their pH optima for activity and location ( Li CM.
  • acid ceramidase (ASAH1, NM_177924.3 or Q13510), neutral ceramidase (ASAH2, NM_019893 or Q9NR71) and alkaline ceramidase (ASAH3, NM_133492, Q8TDN7 or Q5QJU3).
  • a second acid ceramidase (known as acid ceramidase-like or ASAHL) polypeptide has also been described (UniProt Accession number is Q02083).
  • the inhibitors for use in the present invention are those which inhibit at least acid ceramidase and/or the acid ceramidase-like protein, since none of the other two ceramidases show a significative increase in expression in ChoK inhibitor-resistant cells.
  • Acid ceramidase activity is aberrantly expressed in several human cancers. This enzyme may be useful as a new target in cancer, and could be involved in anti-oncogenic treatment resistance ( Seelan RS., Genes Chromosomes Cancer. 2000, 29:137-46 ; Liu X., Front Biosci. 2008;13:2293-8 and Morales A., Oncogene. 2007, 26:905-16 ).
  • Choline kinase inhibitors relates to any compound capable of causing a decrease in the ChoK activity, including those compounds which prevent expression of the ChoK gene, leading to reduced ChoK mRNA or protein levels as well as compounds that inhibit ChoK causing a decrease in the activity of the enzyme.
  • the choline kinase inhibitors are specific for choline kinase alpha.
  • Compounds leading to reduced ChoK mRNA levels can be identified using standard assays for determining mRNA expression levels such as RT-PCR, RNA protection analysis, Northern blot, in situ hybridization, microarray technology and the like.
  • Compounds leading to reduced ChoK protein levels can be identified using standard assays for determining protein expression levels such as Western-blot or Western transfer, ELISA (enzyme-linked immunosorbent assay), RIA (radioimmunoassay), competitive EIA (competitive enzyme immunoassay), DAS-ELISA (double antibody sandwich ELISA), immunocytochemical and immunohistochemical techniques, techniques based on the use of protein biochips or microarrays which include specific antibodies or assays based on colloidal precipitation in formats such as dipsticks.
  • the determination of the inhibitory capacity on the biological activity of choline kinase is detected using standard assays to measure the activity of choline kinase such as the methods based on the detection of the phosphorylation of [ 14 C] labelled choline by ATP in the presence of purified recombinant choline kinase or a fraction enriched in choline kinase followed by detection of the phosphorylated choline using standard analytical techniques (e.g. TLC) as described in EP1710236 .
  • standard analytical techniques e.g. TLC
  • ChoK ⁇ choline kinase alpha
  • choline kinase inhibitors that can be used in the first composition of the present invention are described under I to XVIII in Table 1.
  • Acid ceramidase inhibitors relates to any compound capable of causing a decrease in the acid ceramidase activity, including those compounds which prevent expression of the acid ceramidase gene, leading to reduced acid ceramidase mRNA or protein levels as well as compounds that bind to the active site of acid ceramidase causing a decrease in the activity of the enzyme.
  • Acid ceramidase inhibitors relates to any compound capable of causing a decrease in the acid ceramidase activity, including those compounds which prevent expression of the acid ceramidase gene, leading to reduced acid ceramidase mRNA or protein levels as well as compounds that bind to the active site of acid ceramidase causing a decrease in the activity of the enzyme.
  • Compounds leading to reduced acid ceramidase mRNA levels can be identified using standard assays for determining mRNA expression levels such as RT-PCR, RNA protection analysis, Northern blot, in situ hybridization, microarray technology and the like.
  • Compounds leading to reduced acid ceramidase protein levels can be identified using standard assays for determining protein expression levels such as Western-blot or Western transfer, ELISA (enzyme-linked immunosorbent assay), RIA (radioimmunoassay), competitive EIA (competitive enzyme immunoassay), DAS-ELISA (double antibody sandwich ELISA), immunocytochemical and immunohistochemical techniques, techniques based on the use of protein biochips or microarrays which include specific antibodies or assays based on colloidal precipitation in formats such as dipsticks.
  • standard assays for determining protein expression levels such as Western-blot or Western transfer, ELISA (enzyme-linked immunosorbent assay), RIA (radioimmunoassay), competitive EIA (competitive enzyme immunoassay), DAS-ELISA (double antibody sandwich ELISA), immunocytochemical and immunohistochemical techniques, techniques based on the use of protein biochips or microarrays which include specific antibodies or assays
  • Acid ceramidase inhibitors causing a decrease in the enzymatic activity of acid ceramidase can be identified using standard assays to measure the activity of acid ceramidase using purified acid ceramidase or fractions enriched in acid ceramidase and a substrate thereof. For instance, the method described in ES2233204 which is based on the inhibition of the the hydrolysis of N-(12-(4-nitrobenzene-2-oxa-1,3-diazolo)dodecil) sphingosine (Cer-C12-NBD).
  • Exemplary non-limiting acid ceramidase inhibitors or acid ceramidase-like inhibitors that can be used in the first composition of the present invention are showun under item I to XIII of Table II
  • R is preferably a straight chain alkyl group having 12 to 18 carbon atoms, more preferably a straight chain alkyl group having 13 carbon atoms. Both asymmetric carbon atoms may either take the D or L configuration.
  • R 1 and R 2 may be the same or different and correspond to linear or branched alkyl, or phenylalkyl groups of 1 to 18 carbon atoms having from 0 to several insaturations and substituents (X) at the end of the group wherein X is -OH, -OR wherein R is linear or branched C 1 -C 5 alkyl or metiloxialkyl, -CO 2 H, - CO2R wherein R is as defined previously, -CON(R) 2 wherein R is as defined previously and halogen (F, Cl, Br, I) and R 3 and R 4 are alkyl, alkenyl, aryl, which are the same or different
  • the compound falling under the above general formula suitable for use in the present invention is N-(1-pentyl-2-butyl-3-cyclopro-fenil)ftalamide III
  • R 1 is a linear or branched alkyl, alkenyl, alkinyl, an aryl containing or not heteroatoms and containing substituents in any position or an heterocycle that may be substituted in one or more position
  • Preferred compounds having the above general structure are compunds GT102, GT103 and GT104 as described by Bedia et al. (ChemBioChem., 2007, 8:642-648 ) corresponding to: V
  • Antibodies against an epitope located in either acid ceramidase or in choline kinase alpha isoform may effectively block the function of these proteins and, therefore, can be used as inhibitors in the compositions of the present invention.
  • “Inhibitory antibody”, as used herein, refers to antibodies which are capable of inhibiting at least partially the biological activities of acid ceramidase or of choline kinase alpha isoform actity.
  • the determination of the inhibitory capacity on the biological activity of acid ceramidase is detected using standard assays to measure the activity of acid ceramidase using purified acid ceramidase or fractions enriched in acid ceramidase such as the methods based on the capacity of the antibody of inhibiting the hydrolysis of N-(12-(4-nitrobenzene-2-oxa-1,3-diazolo)dodecil) sphingosine (Cer-C12-NBD) as described e.g. in ES2233204 .
  • the determination of the inhibitory capacity on the biological activity of choline kinase is detected using standard assays to measure the activity of choline kinase such as the methods based on the detection of the phosphorylation of [ 14 C] labelled choline by ATP in the presence of purified recombinant choline kinase or a fraction enriched in choline kinase followed by detection of the phosphorylated choline using standard analytical techniques (e.g. TLC) as described in EP1710236 .
  • standard analytical techniques e.g. TLC
  • Inhibitory antibodies or fragments specific for choline kinase alpha isoform or acid ceramidase may be readily available, or may be readily produced using conventional molecular biology techniques.
  • using immunogens derived from, for example, acid ceramidase or choline kinase alpha isoform it is possible to obtain anti-protein/anti-peptide antisera or monoclonal antibodies by using standard protocols (See, for example, Antibodies: A Laboratory Manual ed. by Harlow and Lane (Cold Spring Harbor Press: 1988 )).
  • a mammal such as a mouse, a hamster or rabbit can be immunized with an immunogenic form of the peptide (e.g., acid ceramidase or choline kinase alpha isoform or an antigenic fragment thereof, which is capable of eliciting an antibody response).
  • an immunogenic form of the peptide e.g., acid ceramidase or choline kinase alpha isoform or an antigenic fragment thereof, which is capable of eliciting an antibody response.
  • Techniques for conferring immunogenicity on a protein or peptide include conjugation to carriers or other techniques, are well known in the art.
  • An immunogenic portion of a polypeptide can be administered in the presence of adjuvant. The progress of immunization can be monitored by detection of antibody titers in plasma or serum. Standard ELISA or other immunoassays can be used with the immunogen as antigen to assess the levels of antibodies.
  • the antibodies forming part of the compositions of the invention are immuno-specific for antigenic determinants of acid ceramidase or choline kinase alpha isoform (or a variant at least 80%, 85%, 90%, 95%, or 98% identical thereto).
  • the immunospecific subject antibodies do not substantially cross react with a non-vertebrate (such as yeast) acid ceramidase or choline kinase-related protein.
  • the antibody has a binding affinity for a non-homologous protein which is at least one order of magnitude, more preferably at least 2 orders of magnitude, and even more preferably at least 3 orders of magnitude less than the binding affinity of the antibody for acid ceramidase or choline kinase.
  • the antibody of the invention is capable of binding an epitope of the choline kinase alpha isoform or of acid ceramidase; typically, at least 6, 8, 10, or 12, contiguous amino acids are required to form an epitope, however, epitopes which involve non-contiguous amino acids may require more, e.g., at least 15, 25, or 50 amino acid.
  • antibody of the invention includes, for example, polyclonal antibodies, monoclonal antibodies, Fab and single chain Fv (scFv) fragments thereof, bispecific antibodies, heteroconjugates, human and humanized antibodies.
  • Such antibodies may be produced in a variety of ways, including hybridoma cultures, recombinant expression in bacteria or mammalian cell cultures, and recombinant expression in transgenic animals. Also antibodies can be produced by selecting a sequence from a library of sequences expressed in display systems such as filamentous phage, bacterial, yeast or ribosome. There is abundant guidance in the literature for selecting a particular production methodology, e.g., Chadd and Chamow, Curr. Opin. BiotechnoL, 12:188-194 (2001 ). The choice of manufacturing methodology depends on several factors including the antibody structure desired, the importance of carbohydrate moieties on the antibodies, ease of culturing and purification, and cost.
  • antibody structures may be generated using standard expression technology, including full-length antibodies, antibody fragments, such as Fab and Fv fragments, as well as chimeric antibodies comprising components from different species.
  • Antibody fragments of small size, such as Fab and Fv fragments, having no effector functions and limited pharmokinetic activity may be generated in a bacterial expression system. Single chain Fv fragments show low immunogenicity and are cleared rapidly from the blood.
  • the antibodies of the invention may be polyclonal antibodies.
  • Such polyclonal antibodies can be produced in a mammal, such as a non-human mammal, for example, following one or more injections of an immunizing agent, and preferably, an adjuvant.
  • the immunizing agent and/or adjuvant will be injected into the mammal by a series of subcutaneous or intraperitoneal injections.
  • the immunizing agent may include choline kinase alpha isoform or acid ceramidase or fragments thereof or a fusion protein thereof or a cell expressing either choline kinase or acid ceramidase.
  • Such proteins, fragments or preparations are introduced into the non-human mammal in the presence of an appropriate adjuvant.
  • an immunogen is as a trasmembrane protein in the surface of a cell (methods described in, e.g., Spiller et al. J. Immunol. Methods, 224: 51-60 (1999 )).
  • These cells can be either cells which naturally express the antigen or in which this expression can be obtained after transfecting the cell with a DNA construct that contains among other DNA sequences those coding the antigen, those necessary for its sufficient expression in the cell. This approach is possible not only when the cell membrane is the natural site in which the antigen is expressed even the antigen once synthesized in the cell is directed at these location by a signal peptide which is added at the antigen coding sequence.
  • the serum contains polyclonal antibodies to undesired epitopes, the polyclonal antibodies can be purified by immunoaffinity chromatography.
  • said antibodies may be monoclonal antibodies.
  • Monoclonal antibodies may be produced by hybridomas, wherein a mouse, hamster, or other appropriate host animal, is immunized with an immunizing agent to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the immunizing agent, e.g. Kohler and Milstein, Nature 256:495 (1975 ).
  • the immunizing agent will typically include a choline kinase alpha isoform, acid ceramidase or a receptor or a fragment thereof or a fusion protein thereof and optionally a carrier or a crude protein preparation which has been enriched for a choline kinase alpha isoform or acid ceramidase or a cell expressing any of said proteins.
  • Such proteins, fragments or preparations are introduced into the non-human mammal in the presence of an appropriate adjuvant.
  • Other form of administration of an immunogen is as a trasmembrane protein in the surface of a cell (methods described in, e.g., Spiller et al. J. Immunol. Methods, 224: 51-60 (1999 )).
  • lymphocytes may be immunized in vitro. Generally, spleen cells or lymph node cells are used if non-human mammalian sources are desired, or peripheral blood lymphocytes ("PBLs”) are used if cells of human origin are desired.
  • PBLs peripheral blood lymphocytes
  • the lymphocytes are fused with an immortalized cell line using a suitable fusing agent, such as polyethylene glycol, to produce a hybridoma cell.
  • a suitable fusing agent such as polyethylene glycol
  • immortalized cell lines are myeloma cells of rat, mouse, bovine or human origin.
  • the hybridoma cells are cultured in a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of unfused, immortalized cells.
  • Clones are isolated using the limiting dilution method and the culture medium (supernatant) in which the hybridoma cells are cultured can be assayed for the presence of monoclonal antibodies directed against ChoK alpha isoform by conventional techniques, such as by flow cytometry or by immunoprecipitation or by other in vitro binding assay, such as RIA or ELISA. Clones can also be cultured in vivo as ascites tumours in an animal.
  • the binding specificity of monoclonal antibodies produced by a clone of hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA) or by immunofluorescent techniques such as fluorescence microscopy or flow cytometry.
  • the monoclonal antibodies secreted by the subclones are suitably separated from the culture medium, ascites fluid, or serum by conventional immunoglobulin purification procedures such as, for example, protein A-Sepharose, hydroxylapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography.
  • the monoclonal antibodies may also be made by recombinant DNA methods, such as those described in US 4,816,567 .
  • DNA encoding the monoclonal antibodies of the invention can be isolated from the choline kinase alpha isoform or acid ceramidase receptor-specific hybridoma cells and sequenced by using conventional procedures, e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies.
  • the hybridoma cells serve as a preferred source of such DNA.
  • the DNA may be inserted into an expression vector, which is then transfected into host cells such as simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells.
  • the DNA also may be modified, for example, by substituting the coding sequence for the murine heavy and light chain constant domains for the homologous human sequences, or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non-immunoglobulin polypeptide.
  • the non-immunoglobulin polypeptide can be substituted for the constant domains of an antibody of the invention, or can be substituted for the variable domains of one antigen-combining site of an antibody of the invention to create a chimeric bivalent antibody.
  • Another method of generating specific antibodies, or antibody fragments, reactive against a target molecule is to screen expression libraries encoding immunoglobulin genes, or portions thereof, expressed in bacteria, yeast, filamentous phages, ribosomes or ribosomal subunits and other display systems. These methods normally use large libraries of antibody sequences or antibody fragment sequences obtained from diverse sources such healthy donors or patients or animals healthy or not. These sequences are cloned and expressed in an appropriate system and selected by its binding affinity for the antigen. Diverse approaches have been described to select antibodies or fragments with desired properties e.g. neutralizing, agonist, etc ( Fernández, Curr. Op. Biotech., 15: 364-373 (2004 ); Schmidt, Eur. J.
  • antibodies and antibody fragments characteristic of hybridomas of the invention can also be produced by recombinant means by extracting messenger RNA, constructing a cDNA library, and selecting clones which encode segments of the antibody molecule.
  • the antibodies may also be engineered to alter its clinical uses.
  • Numerous approaches make use of the molecular biology and genetic techniques such as the good knowledge of the genetics ad structure of the immunoglobulins to construct different modifications of immunoglobulin molecule with the aim of improve its properties for clinical or other uses. Some of them tend to reduce the immunogenicity of the molecule in the species in which should be used and the resultant molecule has a sequence more homologous with this species.
  • Various methods have been used to obtain mAbs of human origin avoiding the non ethically admissible proceedings in healthy humans.
  • the molecular weigh an size are reduced e.g. in order of improving the distribution of the molecule into solid tumours.
  • RNAi Acid ceramidase- or choline kinase-specific RNA interference
  • the inhibitors of the acid ceramidase or choline kinase alpha isoform that form part of the compositions of the invention are RNAi which are capable of knocking down the expression of acid ceramidase and/or choline kinase alpha isoform or any component gene necessary for acid ceramidase and/or choline kinase alpha isoform function.
  • RNAi is a process of sequence-specific post-transcriptional gene repression which can occur in eukaryotic cells. In general, this process involves degradation of an mRNA of a particular sequence induced by double-stranded RNA (dsRNA) that is homologous to that sequence.
  • RNAi may be effected by introduction or expression of relatively short homologous dsRNAs. Indeed, the use of relatively short homologous dsRNAs may have certain advantages as discussed below.
  • the double stranded oligonucleotides used to effect RNAi are preferably less than 30 base pairs in length and, more preferably, comprise about 25, 24, 23, 22, 21, 20, 19, 18 or 17 base pairs of ribonucleic acid.
  • the dsRNA oligonucleotides of the invention may include 3' overhang ends.
  • Exemplary 2-nucleotide 3' overhangs may be composed of ribonucleotide residues of any type and may even be composed of 2'-deoxythymidine residues, which lowers the cost of RNA synthesis and may enhance nuclease resistance of siRNAs in the cell culture medium and within transfected cells (see Elbashir et al., Nature 411: 494-8, 2001 ).
  • dsRNAs Longer dsRNAs of 50, 75, 100 or even 500 base pairs or more may also be utilized in certain embodiments of the invention.
  • Exemplary concentrations of dsRNAs for effecting RNAi are about 0.05 nM, 0.1 nM, 0.5 nM, 1.0 nM, 1.5 nM, 25 nM or 100 nM, although other concentrations may be utilized depending upon the nature of the cells treated, the gene target and other factors readily discernable to the skilled artisan.
  • Exemplary dsRNAs may be synthesized chemically or produced in vitro or in vivo using appropriate expression vectors.
  • Exemplary synthetic RNAs include 21 nucleotide RNAs chemically synthesized using methods known in the art (e.g., Expedite RNA phosphoramidites and thymidine phosphoramidite (Proligo, Germany). Synthetic oligonucleotides are preferably deprotected and gel-purified using methods known in the art (see, e.g., Elbashir et al., Genes Dev. 15: 188-200, 2001 ). Longer RNAs may be transcribed from promoters, such as T7 RNA polymerase promoters, known in the art.
  • RNA target placed in both possible orientations downstream of an in vitro promoter, will transcribe both strands of the target to create a dsRNA oligonucleotide of the desired target sequence.
  • Any of the above RNA species will be designed to include a portion of nucleic acid sequence represented in a target nucleic acid, such as, for example, a nucleic acid that hybridizes, under stringent and/or physiological conditions, to the polynucleotide encoding human acid ceramidase or human ChoK alpha isoform.
  • the specific sequence utilized in design of the oligonucleotides may be any contiguous sequence of nucleotides contained within the expressed gene message of the target. Programs and algorithms, known in the art, may be used to select appropriate target sequences. In addition, optimal sequences may be selected utilizing programs designed to predict the secondary structure of a specified single stranded nucleic acid sequence and allowing selection of those sequences likely to occur in exposed single stranded regions of a folded mRNA. Methods and compositions for designing appropriate oligonucleotides may be found, for example, in U.S. Pat. No. 6,251,588 , the contents of which are incorporated herein by reference.
  • RNA messenger RNA
  • mRNA messenger RNA
  • Secondary structure elements in RNA are formed largely by Watson-Crick type interactions between different regions of the same RNA molecule.
  • Important secondary structural elements include intramolecular double stranded regions, hairpin loops, bulges in duplex RNA and internal loops.
  • Tertiary structural elements are formed when secondary structural elements come in contact with each other or with single stranded regions to produce a more complex three dimensional structure.
  • RNA duplex structures A number of researchers have measured the binding energies of a large number of RNA duplex structures and have derived a set of rules which can be used to predict the secondary structure of RNA (see, e.g., Jaeger et al., Proc. Natl. Acad. Sci. USA 86: 7706, 1989 ; and Turner et al., Annu. Rev. Biophys. Biophys. Chem. 17:167, 1988 ).
  • the rules are useful in identification of RNA structural elements and, in particular, for identifying single stranded RNA regions which may represent preferred segments of the mRNA to target for silencing RNAi, ribozyme or antisense technologies. Accordingly, preferred segments of the mRNA target can be identified for design of the RNAi mediating dsRNA oligonucleotides as well as for design of appropriate ribozyme and hammerhead ribozyme compositions of the invention.
  • siRNA small interfering RNA
  • silencing RNA are a class of 20-25 nucleotide-long double-stranded RNA molecules that play a variety of roles in biology. Most notably, siRNA is involved in the RNA interference (RNAi) pathway where the siRNA interferes with the expression of a specific gene. In addition to their role in the RNAi pathway, siRNAs also act in RNAi-related pathways, e.g., as an antiviral mechanism or in shaping the chromatin structure of a genome. Synthetic siRNAs have been shown to be able to induce RNAi in mammalian cells. This discovery led to a surge in the use of siRNA/RNAi for biomedical research and drug development.
  • MicroRNA are a related class of gene regulatory small RNAs, typically 21-23 nt in length. They typically differ from siRNA because they are processed from single stranded RNA precursors and show only partially complementary to mRNA targets.
  • Initial studies have indicated that miRNAs regulate gene expression post-transcriptionally at the level of translational inhibition at P-Bodies in the cytoplasm.
  • miRNAs may also guide mRNA cleavage similar to siRNAs. This is often the case in plants where the target sites are typically highly complementary to the miRNA. While target sites in plant mRNAs can be found in the 5'UTR, open-reading frames and 3'UTR, in animals, it is the 3' UTR that is the main target.
  • Short hairpin RNA is yet another type of RNA that may be used to effect RNAi. It is a sequence of RNA that makes a tight hairpin turn that can be used to silence gene expression. shRNA is transcribed by RNA polymerase III.
  • RNAi Codex which consists of a database of shRNA related information and an associated website, has been developed as a portal for publicly available shRNA resources and is accessible at http://codex.cshl.org. RNAi Codex currently holds data from the Hannon-Elledge shRNA library and allows the use of biologist-friendly gene names to access information on shRNA constructs that can silence the gene of interest.
  • compositions of the invention comprise ribozymes specifically directed to the mRNA acid ceramidase and/or choline alpha isoform kinase.
  • Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA.
  • the mechanism of ribozyme action involves sequence specific hybridization of the ribozyme molecule to complementary target RNA, followed by an endonucleolytic cleavage event.
  • the composition of ribozyme molecules preferably includes one or more sequences complementary to a target mRNA, and the well known catalytic sequence responsible for mRNA cleavage or a functionally equivalent sequence (see, e.g., U.S. Pat. No. 5,093,246 , incorporated herein by reference in its entirety).
  • ribozymes that cleave mRNA at site specific recognition sequences can be used to destroy target mRNAs
  • the use of hammerhead ribozymes is preferred.
  • Hammerhead ribozymes cleave mRNAs at locations dictated by flanking regions that form complementary base pairs with the target mRNA.
  • the target mRNA has the following sequence of two bases: 5'-UG-3'.
  • the construction and production of hammerhead ribozymes is well known in the art and is described more fully in Haseloff and Gerlach, Nature 334: 585-591, 1988 ; and see PCT Appln. No. WO89/05852 , the contents of which are incorporated herein by reference.
  • Hammerhead ribozyme sequences can be embedded in a stable RNA such as a transfer RNA (tRNA) to increase cleavage efficiency in vivo ( Perriman et al., Proc. Natl. Acad. Sci. USA, 92: 6175-79, 1995 ; de Feyter, and Gaudron, Methods in Molecular Biology, Vol. 74, Chapter 43, "Expressing Ribozymes in Plants,” Edited by Turner, P. C, Humana Press Inc., Totowa, N.J .).
  • tRNA transfer RNA
  • RNA polymerase III-mediated expression of tRNA fusion ribozymes are well known in the art (see, Kawasaki et al., Nature 393: 284-9, 1998 ; Kuwabara et al., Nature Biotechnol. 16: 961-5, 1998 ; and Kuwabara et al., Mol. Cell. 2: 617-27, 1998 ; Koseki et al., J Virol 73: 1868-77, 1999 ; Kuwabara et al., Proc Natl Acad Sci USA 96: 1886-91, 1999 ; Tanabe et al., Nature 406: 473-4, 2000 ).
  • ribozyme cleavage sites within a given target CDNA sequence.
  • the ribozyme is engineered so that the cleavage recognition site is located near the 5' end of the target mRNA--to increase efficiency and minimize the intracellular accumulation of non-functional mRNA transcripts.
  • the use of any cleavage recognition site located in the target sequence encoding different portions of the C-terminal amino acid domains of, for example, long and short forms of target would allow the selective targeting of one or the other form of the target, and thus, have a selective effect on one form of the target gene product.
  • Gene targeting ribozymes necessarily contain a hybridizing region complementary to two regions, each of at least 5 and preferably each 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 contiguous nucleotides in length of a target mRNA, such as an mRNA of a sequence represented in the acid ceramidase or in the choline kinase genes.
  • ribozymes possess highly specific endoribonuclease activity, which autocatalytically cleaves the target sense mRNA.
  • the present invention extends to ribozymes which hybridize to a sense mRNA encoding a target gene such as a therapeutic drug target candidate gene, thereby hybridizing to the sense mRNA and cleaving it, such that it is no longer capable of being translated to synthesize a functional polypeptide product.
  • the ribozymes used in the compositions of the present invention also include RNA endoribonucleases (hereinafter "Cech-type ribozymes”) such as the one which occurs naturally in Tetrahymena thermophila (known as the IVS, or L-19 IVS RNA) and which has been extensively described by Thomas Cech and collaborators ( Zaug et al., Science 224:574-578, 1984 ; Zaug et al., Science 231: 470-475, 1986 ; Zaug et al., Nature 324: 429-433, 1986 ; published International patent application No. WO88/04300 by University Patents Inc.; Been, et al., Cell 47: 207-216, 1986 ).
  • Cech-type ribozymes RNA endoribonucleases
  • the Cech-type ribozymes have an eight base pair active site which hybridizes to a target RNA sequence whereafter cleavage of the target RNA takes place.
  • the invention encompasses those Cech-type ribozymes which target eight base-pair active site sequences that are present in a target gene or nucleic acid sequence.
  • Ribozymes can be composed of modified oligonucleotides (e.g., for improved stability, targeting, etc.) and should be delivered to cells which express the target gene in vivo.
  • a preferred method of delivery involves using a DNA construct "encoding" the ribozyme under the control of a strong constitutive pol III or pol II promoter, so that transfected cells will produce sufficient quantities of the ribozyme to destroy endogenous target messages and inhibit translation. Because ribozymes, unlike antisense molecules, are catalytic, a lower intracellular concentration is required for efficiency.
  • a ribozyme may be designed by first identifying a sequence portion sufficient to cause effective knockdown by RNAi. The same sequence portion may then be incorporated into a ribozyme.
  • the gene-targeting portions of the ribozyme or RNAi are substantially the same sequence of at least 5 and preferably 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 or more contiguous nucleotides of a target nucleic acid, such as a nucleic acid of any of the human acid ceramidase or choline kinase sequences.
  • the method of the invention provides for the use of such methods to select preferred segments of a target mRNA sequence that are predicted to be single-stranded and, further, for the opportunistic utilization of the same or substantially identical target mRNA sequence, preferably comprising about 10-20 consecutive nucleotides of the target mRNA, in the design of both the RNAi oligonucleotides and ribozymes of the invention.
  • a further aspect of the invention relates to the use of the isolated "antisense" nucleic acids to inhibit expression, e.g., by inhibiting transcription and/or translation of acid ceramidase and/or choline kinase alpha isoform nucleic acids.
  • the antisense nucleic acids may bind to the potential drug target by conventional base pair complementarity, or, for example, in the case of binding to DNA duplexes, through specific interactions in the major groove of the double helix. In general, these methods refer to the range of techniques generally employed in the art, and include any methods that rely on specific binding to oligonucleotide sequences.
  • An antisense construct of the present invention can be delivered, for example, as an expression plasmid which, when transcribed in the cell, produces RNA which is complementary to at least a unique portion of the cellular mRNA which encodes a ChoK polypeptide or an acid ceramidase polypeptide.
  • the antisense construct is an oligonucleotide probe, which is generated ex vivo and which, when introduced into the cell causes inhibition of expression by hybridizing with the mRNA and/or genomic sequences of a target nucleic acid.
  • oligonucleotide probes are preferably modified oligonucleotides, which are resistant to endogenous nucleases, e.g., exonucleases and/or endonucleases, and are therefore stable in vivo.
  • exemplary nucleic acid molecules for use as antisense oligonucleotides are phosphoramidate, phosphothioate and methylphosphonate analogs of DNA (see also U.S. Pat. Nos. 5,176,996 ; 5,264,564 ; and 5,256,775 ).
  • oligodeoxyribonucleotides derived from the translation initiation site, e.g., between the -10 and +10 regions of the target gene, are preferred.
  • Antisense approaches involve the design of oligonucleotides (either DNA or RNA) that are complementary to mRNA encoding the target polypeptide. The antisense oligonucleotides will bind to the mRNA transcripts and prevent translation. Absolute complementarity, although preferred, is not required. In the case of double-stranded antisense nucleic acids, a single strand of the duplex DNA may thus be tested, or triplex formation may be assayed.
  • the ability to hybridize will depend on both the degree of complementarity and the length of the antisense nucleic acid. Generally, the longer the hybridizing nucleic acid, the more base mismatches with an RNA it may contain and still form a stable duplex (or triplex, as the case may be). One skilled in the art can ascertain a tolerable degree of mismatch by use of standard procedures to determine the melting point of the hybridized complex.
  • Oligonucleotides that are complementary to the 5' end of the mRNA should work most efficiently at inhibiting translation.
  • sequences complementary to the 3' untranslated sequences of mRNAs have recently been shown to be effective at inhibiting translation of mRNAs as well ( Wagner, Nature 372: 333, 1994 ). Therefore, oligonucleotides complementary to either the 5' or 3' untranslated, non-coding regions of a gene could be used in an antisense approach to inhibit translation of that mRNA.
  • Oligonucleotides complementary to the 5' untranslated region of the mRNA should include the complement of the AUG start codon.
  • Antisense oligonucleotides complementary to mRNA coding regions are less efficient inhibitors of translation but could also be used in accordance with the invention. Whether designed to hybridize to the 5', 3' or coding region of mRNA, antisense nucleic acids should be at least six nucleotides in length, and are preferably less that about 100 and more preferably less than about 50, 25, 17 or 10 nucleotides in length.
  • in vitro studies are first performed to quantitate the ability of the antisense oligonucleotide to inhibit gene expression. It is preferred that these studies utilize controls that distinguish between antisense gene inhibition and nonspecific biological effects of oligonucleotides. It is also preferred that these studies compare levels of the target RNA or protein with that of an internal control RNA or protein. Results obtained using the antisense oligonucleotide may be compared with those obtained using a control oligonucleotide.
  • control oligonucleotide is of approximately the same length as the test oligonucleotide and that the nucleotide sequence of the oligonucleotide differs from the antisense sequence no more than is necessary to prevent specific hybridization to the target sequence.
  • the antisense oligonucleotides can be DNA or RNA or chimeric mixtures or derivatives or modified versions thereof, single-stranded or double-stranded.
  • the oligonucleotide can be modified at the base moiety, sugar moiety, or phosphate backbone, for example, to improve stability of the molecule, hybridization, etc.
  • the oligonucleotide may include other appended groups such as peptides (e.g., for targeting host cell receptors), or agents facilitating transport across the cell membrane (see, e.g., Letsinger et al., Proc. Natl. Acad. Sci. U.S.A. 86: 6553-6556, 1989 ; Lemaitre et al., Proc.
  • oligonucleotide may be conjugated to another molecule, e.g., a peptide, hybridization triggered cross-linking agent, transport agent, hybridization-triggered cleavage agent, etc.
  • the antisense oligonucleotide may comprise at least one modified base moiety which is selected from the group including but not limited to 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xantine, 4-acetylcytosine, 5-(carboxyhydroxytiethyl) uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D-mannosy
  • the antisense oligonucleotide may also comprise at least one modified sugar moiety selected from the group including but not limited to arabinose, 2-fluoroarabinose, xylulose, and hexose.
  • the antisense oligonucleotide can also contain a neutral peptide-like backbone.
  • Such molecules are termned peptide nucleic acid (PNA)-oligomers and are described, e.g., in Perry-O'Keefe et al., Proc. Natl. Acad. Sci. U.S.A. 93: 14670, 1996 , and in Eglom et al., Nature 365: 566, 1993 .
  • the antisense oligonucleotide comprises at least one modified phosphate backbone selected from the group consisting of a phosphorothioate, a phosphorodithioate, a phosphoramidothioate, a phosphoramidate, a phosphordiamidate, a methylphosphonate, an alkyl phosphotriester, and a formacetal or analog thereof.
  • the antisense oligonucleotide is an alpha-anomeric oligonucleotide.
  • An alpha-anomeric oligonucleotide forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual antiparallel orientation, the strands run parallel to each other ( Gautier et al., Nucl. Acids Res. 15: 6625-6641, 1987 ).
  • the oligonucleotide is a 2'-0-methylribonucleotide ( Inoue et al., Nucl. Acids Res. 15: 6131-6148, 1987 ), or a chimeric RNA-DNA analogue ( Inoue et al., FEBS Lett. 215: 327-330, 1987 ).
  • antisense nucleotides complementary to the coding region of a target mRNA sequence can be used, those complementary to the transcribed untranslated region may also be used.
  • a preferred approach utilizes a recombinant DNA construct in which the antisense oligonucleotide is placed under the control of a strong pol III or pol II promoter.
  • the use of such a construct to transfect target cells will result in the transcription of sufficient amounts of single stranded RNAs that will form complementary base pairs with the endogenous potential drug target transcripts and thereby prevent translation.
  • a vector can be introduced such that it is taken up by a cell and directs the transcription of an antisense RNA.
  • Such a vector can remain episomal or become chromosomally integrated, as long as it can be transcribed to produce the desired antisense RNA.
  • Such vectors can be constructed by recombinant DNA technology methods standard in the art.
  • Vectors can be plasmid, viral, or others known in the art, used for replication and expression in mammalian cells.
  • Expression of the sequence encoding the antisense RNA can be by any promoter known in the art to act in mammalian, preferably human cells. Such promoters can be inducible or constitutive.
  • Such promoters include but are not limited to: the SV40 early promoter region ( Bernoist and Chambon, Nature 290: 304-310, 1981 ), the promoter contained in the 3' long terminal repeat of Rous sarcoma virus ( Yamamoto et al., Cell 22: 787-797, 1980 ), the herpes thymidine kinase promoter ( Wagner et al., Proc. Natl. Acad. Sci. U.S.A. 78: 1441-1445, 1981 ), the regulatory sequences of the metallothionein gene ( Brinster et al, Nature 296: 39-42, 1982 ), etc. Any type of plasmid, cosmid, YAC or viral vector can be used to prepare the recombinant DNA construct, which can be introduced directly into the tissue site.
  • target gene expression can be reduced by targeting deoxyribonucleotide sequences complementary to the regulatory region of the gene (i.e., the promoter and/or enhancers) to form triple helical structures that prevent transcription of the gene in target cells in the body (see generally, Helene, Anticancer Drug Des. 6(6): 569-84, 1991 ; Helene et al., Ann. N.Y. Acad. Sci., 660: 27-36, 1992 ; and Maher, Bioassays 14(12): 807-15, 1992 ).
  • deoxyribonucleotide sequences complementary to the regulatory region of the gene i.e., the promoter and/or enhancers
  • Nucleic acid molecules to be used in triple helix formation for the inhibition of transcription are preferably single stranded and composed of deoxyribonucleotides.
  • the base composition of these oligonucleotides should promote triple helix formation via Hoogsteen base pairing rules, which generally require sizable stretches of either purines or pyrimidines to be present on one strand of a duplex.
  • Nucleotide sequences may be pyrimidine-based, which will result in TAT and CGC triplets across the three associated strands of the resulting triple helix.
  • the pyrimidine-rich molecules provide base complementarity to a purine-rich region of a single strand of the duplex in a parallel orientation to that strand.
  • nucleic acid molecules may be chosen that are purine-rich, for example, containing a stretch of G residues. These molecules will form a triple helix with a DNA duplex that is rich in GC pairs, in which the majority of the purine residues are located on a single strand of the targeted duplex, resulting in CGC triplets across the three strands in the triplex.
  • the potential target sequences that can be targeted for triple helix formation may be increased by creating a so called “switchback" nucleic acid molecule.
  • Switchback molecules are synthesized in an alternating 5'-3',3'-5' manner, such that they base pair with first one strand of a duplex and then the other, eliminating the necessity for a sizable stretch of either purines or pyrimidines to be present on one strand of a duplex.
  • the antisense oligonucleotides are morpholino antisenses.
  • Morpholinos are synthetic molecules which are the product of a redesign of natural nucleic acid structure. Usually 25 bases in length, they bind to complementary sequences of RNA by standard nucleic acid base-pairing. Structurally, the difference between morpholinos and DNA is that while morpholinos have standard nucleic acid bases, those bases are bound to morpholine rings instead of deoxyribose rings, and linked through phosphorodiamidate groups instead of phosphates.
  • Morpholinos are not chimeric oligos; the entire backbone of a morpholino is made from these modified subunits. Morpholinos are most commonly used as single-stranded oligos, though heteroduplexes of a morpholino strand and a complementary DNA strand may be used in combination with cationic cytosolic delivery reagents.
  • morpholinos do not degrade their target RNA molecules. Instead, morpholinos act by "steric blocking," binding to a target sequence within an RNA and simply getting in the way of molecules which might otherwise interact with the RNA. Morpholino oligos are often used to investigate the role of a specific mRNA transcript in an embryo, such as eggs or embryos of zebrafish, African clawed frog (Xenopus), chick, and sea urchin, producing morphant embryos. With appropriate cytosolic delivery systems, morpholinos are effective in cell culture.
  • morpholinos can interfere with progression of the ribosomal initiation complex from the 5' cap to the start codon. This prevents translation of the coding region of the targeted transcript (called “knocking down” gene expression). Morpholinos provide a convenient means of knocking down expression of the protein and learning how that knockdown changes the cells or organism. Some morpholinos knock down expression so effectively that after degradation of preexisting proteins the targeted proteins become undetectable by Western blot.
  • Morpholinos can also interfere with pre-mRNA processing steps, usually by preventing the splice-directing snRNP complexes from binding to their targets at the borders of introns on a strand of pre-RNA. Preventing U1 (at the donor site) or U2/U5 (at the polypyrimidine moiety and acceptor site) from binding can cause modified splicing, commonly leading to exclusions of exons from the mature mRNA. Targeting some splice targets results in intron inclusions, while activation of cryptic splice sites can lead to partial inclusions or exclusions. Targets of U11/U12 snRNPs can also be blocked. Splice modification can be conveniently assayed by reverse-transcriptase polymerase chain reaction (RT-PCR) and is seen as a band shift after gel electrophoresis of RT-PCR products.
  • RT-PCR reverse-transcriptase polymerase chain reaction
  • Morpholinos have also been used to block miRNA activity, ribozyme activity, intronic splice silencers, and splice enhancers. U2 and U12 snRNP functions have been inhibited by Morpholinos. Morpholinos targeted to "slippery" mRNA sequences within protein coding regions can induce translational frameshifts. Activities of Morpholinos against this variety of targets suggest that Morpholinos can be used as a general-purpose tool for blocking interactions of proteins or nucleic acids with mRNA.
  • a further aspect of the invention relates to compositions wherein the acid ceramidase inhibitor and/or the choline kinase alpha isoform inhibitor is/are DNA enzymes.
  • DNA enzymes incorporate some of the mechanistic features of both antisense and ribozyme technologies. DNA enzymes are designed so that they recognize a particular target nucleic acid sequence, much like an antisense oligonucleotide, however much like a ribozyme they are catalytic and specifically cleave the target nucleic acid.
  • the 10-23 DNA enzyme comprises a loop structure which connect two arms.
  • the two arms provide specificity by recognizing the particular target nucleic acid sequence while the loop structure provides catalytic function under physiological conditions.
  • the unique or substantially sequence is a G/C rich of approximately 18 to 22 nucleotides. High G/C content helps insure a stronger interaction between the DNA enzyme and the target sequence.
  • the specific antisense recognition sequence that will target the enzyme to the message is divided so that it comprises the two arms of the DNA enzyme, and the DNA enzyme loop is placed between the two specific arms.
  • DNA enzymes can be found, for example, in U.S. Pat. No. 6,110,462 .
  • methods of delivery DNA ribozymes in vitro or in vivo include methods of delivery RNA ribozyme, as outlined in detail above.
  • DNA enzymes can be optionally modified to improve stability and improve resistance to degradation.
  • Antisense RNA and DNA, ribozyme, RNAi and triple helix molecules of the invention may be prepared by any method known in the art for the synthesis of DNA and RNA molecules. These include techniques for chemically synthesizing oligodeoxyribonucleotides and oligoribonucleotides well known in the art such as for example solid phase phosphoramidite chemical synthesis. Alternatively, RNA molecules may be generated by in vitro and in vivo transcription of DNA sequences encoding the antisense RNA molecule. Such DNA sequences may be incorporated into a wide variety of vectors which incorporate suitable RNA polymerase promoters such as the T7 or SP6 polymerase promoters.
  • antisense cDNA constructs that synthesize antisense RNA constitutively or inducibly, depending on the promoter used, can be introduced stably into cell lines.
  • various well-known modifications to nucleic acid molecules may be introduced as a means of increasing intracellular stability and half-life. Possible modifications include but are not limited to the addition of flanking sequences of ribonucleotides or deoxyribonucleotides to the 5' and/or 3' ends of the molecule or the use of phosphorothioate or 2' O-methyl rather than phosphodiesterase linkages within the oligodeoxyribonucleotide backbone.
  • ChoK alpha isoform inhibitors and acid ceramidase inhibitors include MN58b (Structure provided in line II in Table 1) and NOE (structure provided in line 6, table 2), RSM-932A (structure provided in line I in table 1) and NOE, MN58b and D-NMAPPD (structure provided in line VIII in table 2) and RSM-932A and D-NMAPPD.
  • the authors of the present invention have found that, surprisingly, the resistance to the treatment with the ChoK inhibitor MN58b does not correlate with resistances to conventional chemotherapeutic agentes such as cisplatin, taxol, virelbine or gemcitabine. This has been observed both in tumours resistant to ChoK inhibitors (see example 1) as well as in established tumor cell lines selected by repeated cycles of growth in the presence of a ChoK inhibitor (see example 6 of the invention). Without wishing to be bound by any theory, it is believed that the non-crossed resistance between ChoK inhibitors and cisplatin is due to the fact that both drugs act through different mechanisms.
  • the invention relates to a composition (hereinafter second composition of the invention) comprising, separately or together, a first component comprising one or more ChoK inhibitors specific for the choline kinase alpha isoform and a second component comprising one or more chemotherapy agents, wherein the chemotherapy agent is selected from the group consisting of a DNA-alkylating agent, an antimetabolite, a mitotic inhibitor, an anthracycline, a topoisomerase I inhibitor, a topoisomerase II inhibitor, cetuximab, gefitinib and imatinib.
  • composition refers to one or more compounds in various combinations according to alternative embodiments of this invention.
  • the composition comprises at least a ChoK inhibitor and at least an alkylating agent.
  • ChoK inhibitors suitable for use in the compositions of the invention include any of the ChoK inhibitors defined previously in Table 1 as forming part of the first composition of the invention.
  • the ChoK inhibitor is an inhibitor specific for ChoK alpha isoform.
  • chemotherapy agent refers to a chemical agent that inhibits the proliferation, growth, life-span or metastatic activity of cancer cells and include, without limitation, DNA-alkylating drugs, antimetabolites, mitotic inhibitors, anthracyclines, topoisomerase I and II inhibitors, cetuximab, gefitinib and imatinib.
  • the chemotherapy agent is an alkylating agent and, more in particular, a DNA-alkylating agent or an antimetabolite.
  • alkylating agent relate to compounds capable of adding alkyl residues to the genetic material of rapidly dividing cells thus leading to replication arrest and cell death.
  • agents include platinum-based compounds, nitrogen mustards, nitrosoureas, ethylenimine derivatives, alkyl sulfonates, and triazenes, including, but not limited to, mechlorethamine, cyclophosphamide (CytoxanTM), melphalan (L-sarcolysin), etoposide, carmustine (BCNU), lomustine (CCNU), semustine (methyl-CCNU), streptozocin, chlorozotocin, uracil mustard, chlormethine, ifosfamide, chlorambucil, pipobroman, triethylenemelamine, triethylenethiophosphoramine, busulfan, procarbazine, dacarbazine, and temozolomide.
  • the alkylating agent is a platinum-based compound.
  • platinum-based compounds that can be used in the present invention include, without limitation, cisplatin, carboplatin, iproplatin, tetraplatin, oxaliplatin, JM118, JM149, JM216, JM335, transplatino, cis, trans, cis-Pt(NH3)(C6H11NH2)(OOCC3H7)2Cl, nedaplatin, malanate-1,2-diaminociclohexanoplatin(II), 5-sulphosalycilate-trans-(1,2-diaminociclohexane)platin (II) (SSP), poly-[(trans-1,2-diaminocyclohexane)platin]-carboxyamilose (POLY-PLAT) and 4-hydroxy-sulphonylphenylacetate (trans-1,2-diaminocyclohexane) plat
  • antimetabolite relates, in a broad sense, to substances which disturb normal metabolism and substances which inhibit the electron transfer system to prevent the production of energy-rich intermediates, due to their structural or functional similarities to metabolites that are important for living organisms (such as vitamins, coenzymes, amino acids and saccharides).
  • antimetabolites that have antitumor activities include but are not limited to folic acid analogs (e.g., methotrexate (amethopterin)), denopterin, edatrexate, methotrexate, nolatrexed, pemetrexed, piritrexim, pteropterin, raltitrexed, trimetrexate; pyrimidine analogs (e.g., fluorouracil (5- fluorouracil; 5-FU(R)), floxuridine (fluorode-oxyuridine; FudR), doxifluridine and cytarabine (cytosine arabinoside)); purine analogs (e.g., mercaptopurine (6-mercaptopurine; 6-MP), thioguanine (6-thioguanine; TG), and pentostatin (2'-deoxycoformycin)).
  • folic acid analogs e.g., methotrexate (amethopterin
  • Preferred combinations include, without limitation, MN58b and cisplatin and RSM-932A and cisplatin, MN58b and 5-fluorouracile and RSM-932A and 5-fluorouracile.
  • example 8 of the present invention shows that treatment of tumor cells with a combination of a death receptor ligand and an inhibitor of ChoK results in an increased inhibition of cell proliferation when compared with the treatment with the death receptor ligand or the ChoK inhibitor separately.
  • the treatment of colon cancer cells with a combination of the choline kinase inhibitor RSM-932A (ChoKI) and TRAIL results in an increased cytotoxicity when compared to the treatment of the same cells with each compound taken individually.
  • example 8 of the present invention shows that the combined use of the ChoK inhibitor MN58b and TRAIL results in an inhibition of tumor growth in a tumor xenograft model which is higher than that observed when each of the compounds is used separately.
  • composition comprising, together or separately, a first component comprising one or more inhibitors of ChoK and a second component which is one or more death receptor ligands.
  • ChoK inhibitors suitable for use in the compositions include any of the ChoK inhibitors defined previously in Table 1 as forming part of the first composition of the invention.
  • the ChoK inhibitors is an inhibitor specific for ChoK ⁇ .
  • Death receptor ligands suitable for use in the compositions include NGF, CD40L, CD137L/4-1BBL, TNF- ⁇ CD134L/OX40L, CD27L/CD70, FasL/CD95, CD30L, TNF- ⁇ /LT- a, LT- ⁇ and TRAIL.
  • the TNF family member is TRAIL, a functionally equivalent derivative thereof or a small mimic compound thereof.
  • TRAIL TNF-related apoptosis inducing ligand
  • Apo-2 ligand also known as "Apo-2 ligand", “Apo-2L”, “Apo2L”, “Apo2L/TRAIL” and "Apo-2 ligand/TRAIL”
  • TRAIL was identified several years ago as a member of the TNF family of cytokines, ( Pitti et al., 1996, J.Biol.Chem., 271:12687-12690 and US Patent 6,284,236 ).
  • the full-length native sequence human TRAIL polypeptide is a 281 amino acid long, Type II transmembrane protein having a sequence as defined in SEQ ID NO: 7 (UniProt accession P50591). Crystallographic studies of soluble forms of TRAIL reveal a homotrimeric structure similar to the structures of TNF and other related proteins. TRAIL, unlike other TNF family members however, was found to have a unique structural feature in that three cysteine residues (at position 230 of each subunit in the homotrimer) together coordinate a zinc atom, and that the zinc binding is important for trimer stability and biological activity.
  • the present invention contemplates the use of any of the three different TRAIL isoforms (TRAIL ⁇ , TRAIL ⁇ and TRAIL ⁇ ) o combinations thereof.
  • TRAIL variants include soluble TRAIL isoforms such as those described in WO08088582 and US6284236 or the TRAIL fragments 95-281, 114-281 described in US2002128438 , scFv:sTRAIL fusions as described by Bremer et al (Neoplasia, 2004, 6:636-45 ), alternatively-spliced forms of TRAIL as described in US2002061525 , TRAIL-receptor binding peptides as described in WO04101608 , TRAIL variants with increased specificity for the pro-apoptotic receptors such as the 19 IL, 199V, 20 1 R, 213W, 215D and/or 193S TRAIL mutants as described in WO07063301 or variants selected by phage-display on receptors as described in WO04001009A , agonistic antibodies directed against TRAIL-cognate receptors TRAIL-R1 (DR4) and
  • Small molecule TRAIL mimics having pro-apoptotic effect include the compounds described in WO2008094319 .
  • Preferred combinations include, without limitation, RSM-932A and the extracellular region of human TRAIL (amino acids 95-281) and MN58b and the extracellular region of human TRAIL (amino acids 95-281).
  • the compounds that form part of the first and second compositions of the invention include not only the compounds as such but also pharmaceutically acceptable salts, solvates, prodrugs thereof.
  • pharmaceutically acceptable salts, solvates, prodrugs refers to any pharmaceutically salt, ester, solvate or any other compound which when administered to a receptor is able to provide (directly or indirectly) a compound as described in the present document.
  • pharmaceutically unacceptable salts are also within the scope of the invention because the latter can be useful in the preparation of pharmaceutically acceptable salts.
  • the preparation of salts, prodrugs and derivatives can be carried out by means of methods known in the art.
  • salts of compounds provided in the present document are synthesized by means of conventional chemical methods from an original compound containing a basic or acid residue.
  • Such salts are generally prepared, for example, by reacting the free acid or base forms of the compounds with a stoichiometric amount of the suitable base or acid in water or an organic solvent or a mixture of both.
  • Non-aqueous media such as DMSO (dimethylsulphoxide), ether, ethyl acetate, ethanol, isopropanol or acetonitrile are generally preferred.
  • acid addition salts include mineral acid addition salts such as for example, hydrochloride, bromohydrate, iodohydrate, sulfate, nitrate, phosphate and organic acid addition salts such as for example, acetate, maleate, fumarate, citrate, oxalate, succinate, tartrate, malate, mandelate, methanesulfonate and p-toluenesulfonate.
  • base addition salts include inorganic salts such as for example sodium, potassium, bromide, calcium, ammonium, magnesium, aluminium and lithium salts and organic base salts such as for example ethylenediamine, ethanolamine, N,N-dialkylenethanolamine, triethanolamine, glucamine and basic amino acid salts.
  • the particularly preferred derivatives or prodrugs are those increasing the bioavailability of the compounds of this invention when such compounds are administered to a patient (for example, by making a compound administered orally be absorbed more easily by blood), or enhancing the release of the original compound in a biological compartment (for example, the brain or the lymphatic system) in relation to the original species.
  • the invention also provides compositions wherein at least one of the compounds are found as prodrug.
  • prodrug is used in its widest sense and includes those derivatives which are converted in vivo into the compounds of the invention. Such derivatives are evident for the persons skilled in the art and depending on the functional groups present in the molecule and without limitation, include the following derivatives of the present compounds: esters, amino acid esters, phosphate esters, metal salt sulfonate esters, carbamates and amides. Examples of methods for producing a prodrug of a given active compound are known by the person skilled in the art and can be found for example in Krogsgaard-Larsen et al. "Textbook of Drug design and Discovery” Taylor & Francis (April 2002 ).
  • the compounds of the invention can be in crystalline form as free compounds or as solvates and it is intended that both of them are within the scope of the present invention.
  • the solvation methods are generally known in the art.
  • the suitable solvates are pharmaceutically acceptable solvates.
  • the solvate is a hydrate.
  • the compounds forming the compositions of the invention can include enantiomers, depending on the presence of chiral centers on a C, or isomers, depending on the presence of multiple bonds (for example, Z, E). The individual isomers, enantiomers or diastereoisomers and the mixtures thereof are included within the scope of the present invention.
  • hydroxyl groups act as hydrogen bond donors and intra or intermolecular links can be established even in the case of phenols.
  • the presence of carbonyl or carboxyl groups generates proton acceptor groups in the molecule.
  • the presence of halogens generates very deficient carbons and considerably modifies the biological properties.
  • the amino groups generate good nucleophiles on the molecule and in most cases significantly modify its polarity and polarizability and the presence of additional alkyl and/or aryl groups increases the lypophilicity of the molecules.
  • the invention provides pharmaceutical compositions comprising the first or second compositons of the invention, their pharmaceutically acceptable salt, derivative, prodrug, solvate or stereoisomer thereof together with a pharmaceutically acceptable carrier, adjuvant or vehicle for the administration to a patient.
  • pharmaceutically acceptable carrier means a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject agents from one organ, or portion of the body, to another organ, or portion of the body.
  • Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation.
  • materials which can serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol, solutol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and
  • compositions can be administered by any suitable administration route, for example an oral, topical, rectal or parenteral route (including subcutaneous, intraperitoneal, intradermal, intramuscular and intravenous route).
  • suitable administration route for example an oral, topical, rectal or parenteral route (including subcutaneous, intraperitoneal, intradermal, intramuscular and intravenous route).
  • Suitable pharmaceutical forms for oral administration include any solid composition (tablets, pastilles, capsules, granules, etc.) or liquid composition (solutions, suspensions, emulsions, syrups, etc.) and can contain conventional excipients known in the art, such as binding agents, for example syrup, acacia, gelatin, sorbitol, tragacanth, or polyvinylpyrrolidone; fillers, for example lactose, sugar, cornstarch, calcium fosfate, sorbitol or glycine; lubricants for the preparation of tablets, for example magnesium stearate, disintegrants, for example starch, polyvinylpirrolidone, sodium starch glycolate or microcrystalline cellulose; or pharmaceutically acceptable wetting agents such as sodium laurylsulfate.
  • binding agents for example syrup, acacia, gelatin, sorbitol, tragacanth, or polyvinylpyrrolidone
  • fillers for example lactose,
  • Solid oral compositions can be prepared by means of conventional methods for mixing, filling or preparing tablets.
  • the repeated mixing operations can be used to distribute the active ingredient through the entire compositions by using large amounts of filler agents. Such operations are conventional in the art.
  • the tablets can be prepared, for example by means of wet or dry granulation and can be optionally coated according to methods well known in normal pharmaceutical practice, particularly with an enteric coating.
  • compositions can also be adapted for parenteral administration such as sterile solutions, suspensions or lyophilized products in a suitable unitary pharmaceutical form.
  • Suitable excipients such as bulk agents, buffering agents or surfactants can be used.
  • the mentioned formulations will be prepared using usual methods such as those described or referred to in Spanish Pharmacopoeia and the Pharmacopoeia of the United States and in similar reference texts.
  • the administration of the compounds or compositions used in the present invention can be by any suitable method, such as intravenous infusion, oral preparations and intraperitoneal and intravenous administration. Nevertheless, the preferred administration route will depend on the patient's condition. Oral administration is preferred due to the comfort for the patient and the chronic character of the diseases which are to be treated.
  • compositions of the invention will preferably be found in pharmaceutically acceptable or substantially pure form, i.e. the compositions of the invention have a pharmaceutically acceptable purity level excluding the pharmaceutically acceptable excipients and not including material considered to be toxic at the normal dosage levels.
  • the purity levels for the inhibitors of acid ceramidase or for the inhibitors of choline kinase preferably exceed 50%, more preferably exceed 70%, more preferable exceed 90%. In a preferred embodiment, they exceed 95%.
  • the therapeutically effective amounts of the inhibitors of acid ceramidase or of the chemotherapeutic agent, and of the inhibitors of choline kinase alpha isoform in the compositions of the invention will generally depend, among other factors, on the individual who is to be treated, on the severity of the disease said individual suffers from, on the administration form chosen etc. For this reason, the doses mentioned in this invention must be considered as guides for the person skilled in the art and the latter must adjust the doses according to the variables mentioned previously.
  • an inhibitor of acid ceramidase can be administered once or more times a day, for example, 1, 2, 3 or 4 times a day in a typical daily total amount comprised between 1 and 200 mg/kg body mass/day, preferably 1-10 mg/kg body mass/day.
  • an inhibitor of choline kinase can be administered once or more times a day, for example, 1, 2, 3 or 4 times a day in a typical daily total amount comprised between 1 and 200 mg/kg body mass/day, preferably 1-10 mg/kg body mass/day.
  • compositions according to the present invention can be formulated as a single preparation or, alternatively, they may be provided as a product for the simultaneous, concurrent, separate or sequential administration.
  • compositions described in this invention, their pharmaceutically acceptable salts, prodrugs and/or solvates, as well as the pharmaceutical compositions containing them can be used together with other additional drugs to provide a combination therapy.
  • Said additional drugs can form part of the same pharmaceutical composition or can alternatively be provided in the form of a separate composition for its simultaneous or non-simultaneous administration with the pharmaceutical composition comprising an inhibitor of acid ceramidase and an inhibitor of choline kinase or a pharmaceutically acceptable prodrug, solvate or salt thereof.
  • the other drugs can form part of the same composition or be provided as a separate composition for its administration at the same time or at different times.
  • compositions of the invention can be administered in combination with other chemotherapeutic agents known in the art such as
  • the composition may addditionaly comprise an alkylating agent.
  • alkylating agents for use in the first composition of the invention include platinum-based compounds such as carboplatin, cisplatin, nedaplatin, oxaliplatin, triplatin tetranitrate, satraplatin and combinations thereof; alkyl sulfonates such as Busulfan, ethyleneimines and methylmelamines such as hexamethylmetamine, altretamine or Thiotepa, nitrogen mustards such as cyclophosphamide, mechlorethamine or mustine, uramustine or uracil mustard, Melphalan, Chlorambucil or Ifosfamide, nitrosoureas such as carmustine or streptozocin, triazenes such as dacarbazine and imidazotetrazines such as temozolomide.
  • platinum-based compounds such as carboplatin, cisplatin, neda
  • the compositions may comprise as well a death receptor ligand.
  • said death receptor ligand is selected from the group consisting of NGF, CD40L, CD137L/4-1BBL, TNF- ⁇ CD134L/OX40L, CD27L/CD70, FasL/CD95, CD30L, TNF- ⁇ /LT- a, LT- ⁇ and TRAIL.
  • the TNF family member is TRAIL, a functionally equivalent derivative thereof or a small mimic compound thereof TRAIL (TNF-related apoptosis inducing ligand), also known as "Apo-2 ligand", “Apo-2L”, “Apo2L”, “Apo2L/TRAIL” and "Apo-2 ligand/TRAIL”, is a molecule which is capable of inducing apoptosis in cells expressing the TRAIL cognate receptor.
  • TRAIL was identified several years ago as a member of the TNF family of cytokines, ( Pitti et al., 1996, J.Biol.Chem., 271:12687-12690 and US Patent 6,284,236 ).
  • TRAIL polypeptide The full-length native sequence human TRAIL polypeptide is a 281 amino acid long, Type II transmembrane protein. Crystallographic studies of soluble forms of TRAIL reveal a homotrimeric structure similar to the structures of TNF and other related proteins. TRAIL, unlike other TNF family members however, was found to have a unique structural feature in that three cysteine residues (at position 230 of each subunit in the homotrimer) together coordinate a zinc atom, and that the zinc binding is important for trimer stability and biological activity. The present invention contemplates the use of any of the three different TRAIL isoforms (TRAIL ⁇ , TRAIL ⁇ and TRAIL ⁇ ) o combinations thereof.
  • TRAIL variants include soluble TRAIL isoforms such as those described in WO08088582 and US6284236 or the TRAIL fragments 95-281, 114-281 described in US2002128438 , scFv:sTRAIL fusions as described by Bremer et al (Neoplasia, 2004, 6:636-45 ), alternatively-spliced forms of TRAIL as described in US2002061525 , TRAIL-receptor binding peptides as described in WO04101608 , TRAIL variants with increased specificity for the pro-apoptotic receptors such as the 19 IL, 199V, 201R, 213W, 215D and/or 193S TRAIL mutants as described in WO07063301 or variants selected by phage-display on receptors as described in WO04001009A , agonistic antibodies directed against TRAIL-cognate receptors TRAIL-R1 (DR4) and
  • Small molecule TRAIL mimics having pro-apoptotic effect include the compounds described in WO2008094319 .
  • compositions of the invention in view of their synergistic effect in inhibiting growth of tumor cells, can be used in medicine.
  • the invention relates to the compositions of the invention for use in medicine.
  • the invention relates to the use of a composition
  • a composition comprising (i) an inhibitor of acid ceramidase and an inhibitor of choline kinase specific for the choline kinase alpha isoform or, (ii) an inhibitor of choline kinase specific for the choline kinase alpha isoform and a chemotherapeutic agent, wherein the chemotherapy agent is selected from the group consisting of a DNA-alkylating agent, an antimetabolite, a mitotic inhibitor, an anthracycline, a topoisomerase I inhibitor, a topoisomerase II inhibitor, cetuximab, gefitinib and imatinib, for the manufacture of a medicament for the treatment of cancer.
  • the invention relates to a composition
  • a composition comprising (i) an inhibitor of acid ceramidase and an inhibitor of choline kinase specific for the choline kinase alpha isoform or, (ii) an inhibitor of choline kinase specific for the choline kinase alpha isoform and a chemotherapeutic agent , wherein the chemotherapy agent is selected from the group consisting of a DNA-alkylating agent, an antimetabolite, a mitotic inhibitor, an anthracycline, a topoisomerase I inhibitor, a topoisomerase II inhibitor, cetuximab, gefitinib and imatinib, for use in the treatment of cancer.
  • the cancer is selected from the group consisting of heavy chain disease, leukemias (e.g., acute myeloid leukemia, chronic myeloid leukemia, chronic myelomonocytic leukemia, acute promyelocytic leukemia, myelodysplastic syndrome, juvenile myelomonocytic leukemia, etc.), metastases, neoplasms, tumors (e.g., acoustic neuroma, adenocarcinoma, adrenal cortical cancer, anal carcinoma, angiosarcoma, astrocytoma, basal cell carcinoma, bile duct carcinoma, bladder carcinoma, brain cancer, breast cancer, bronchogenic carcinoma, cancer of the peritoneum, cervical cancer, chondrosarcoma, chordoma, choriocarcinoma, colon carcinoma, colorectal cancer, cr
  • the cancer is lung cancer.
  • "Lung cancer”, as used herein, relates to any neoplastic modification affecting one of more cells present in the lung tissue.
  • Exemplary non-limiting types of lung cancer that can be treated using the compositions of the invention include small and non-small cell lung cancer, including squamous cell carcinoma, adenocarcinoma and large cell carcinoma and mesothelioma.
  • the lung cancer is non-small cell lung cancer.
  • the cancer is colon cancer or colorectal cancer.
  • colon cancer includes any type of colon, rectal and appendix neoplasia and refers both to early and late adenomas and to carcinomas as well as to the hereditary, familial or sporadic cancer.
  • Hereditary CRC includes those syndromes which include the presence of polyps, such as the hamartomatous polyposis syndromes and the most known, familial adenomatous polyposis (FAP) as well as nonpolyposis syndromes such as hereditary nonpolyposis colorectal cancer (HNPCC) or Lynch syndrome I.
  • the present invention allows the diagnosis of colorectal cancer in its different stages such as stages A, B, C1, C2 and D according to Dukes' classification, stages A, B1, B2, B3, C1, C2, C3 and D according to the Astler-Coller classification, stages TX, T0, Tis, T1, T2, T3, NX, N0, N1, N2, MX, M0 and M1 according to the TNM system as well as stages 0, I, II, III and IV according to the AJCC (American Joint Committee on Cancer) classification.
  • stages A, B, C1, C2 and D according to Dukes' classification
  • stages A, B1, B2, B3, C1, C2, C3 and D according to the Astler-Coller classification
  • stages TX, T0, Tis, T1, T2, T3, NX, N0, N1, N2, MX, M0 and M1 according to the TNM system as well as stages 0, I, II, III and IV according to the AJCC (American Joint Committee on Cancer) classification.
  • compositions according to the present invention can be formulated as a single preparation or, alternatively, they may be provided as a product for the simultaneous, concurrent, separate or sequential administration.
  • the authors of the present invention have also observed that the response to inhibitors of choline kinase is enhanced when the cells are treated previously or simultaneously with an acid ceramidase inhibitor (see example 6 of the present invention), with a chemotherapeutic agent (see examples 7 and 9) or with a death receptor ligand (see example 8).
  • a method for increasing the sensitivity of a tumor cells to a choline kinase inhibitor which comprises treating said-tumor cells with an acid ceramidase inhibitor, with a chemotherapeutic agent or with a death receptor ligand.
  • the invention relates to the use of an inhibitor of acid ceramidase, a chemotherapeutic agent or a death receptor ligand to increase the sensitivity of a tumor cell to a choline kinase inhibitor.
  • sensitivity refers to the response of a cell to a given choline kinase inhibitor and is usually determined as the minimal dosis of the choline kinase inhibitor which causes a 50% inhinbition in the growth of a tumor cell.
  • the acid ceramidase inhibitor, with a chemotherapeutic agent or with a death receptor ligand act by increasing the sensitivity or decreasing the minimal dosis needed to achieve 50% growth inhibition.
  • the sensitization of tumor cells to the treatment with ChoK inhibitors can be carried by treating the cells with an acid ceramidase inhibitor, with the chemotherapeutic agent or with the death receptor ligand simultaneously, after or prior to the administration of the ChoK inhibitors.
  • the acid ceramidase inhibitor, the chemotherapeutic agent and the death receptor ligand that can be used to increase the sensitivity of a tumor cell to a ChoK inhibitor can be any of the compounds previously described as forming part of the compositions of the invention.
  • the ChoK inhibitors that can be used for the treatment of cells which have been sensitized with the acid ceramidase inhibitors or with the death receptor ligand are essentially any of the inhibitors previously described as components of the compositions of the invention.
  • the findings of the authors of the invention open the possibility of identifying cancer patients which are likely to show resistance to the treatment with ChoK inhibitors by determining the levels of acid ceramidase in a sample of said patient. If acid ceramidase levels are increased with respect to a reference sample, this will be indicative that the patients are likely to show resistance to ChoK inhibitors since the pro-apoptotic ceramidase produced in response to ChoK inhibition will be hydrolysed giving rise to sphingosine which has pro-mitotic effects. On the contrary, if the levels of acid ceramidase are decreased or at least not increased with respect to the levels in a reference sample, this is indicative that the patient will respond favourably to the treatment with ChoK inhibitors.
  • first method for the identification of cancer patients resistant to therapy with ChoK inhibitors comprising determining the levels of acid ceramidase in a sample from said patient wherein the patient is identified as being resistant to ChoK inhibitors when the acid ceramidase levels in said sample are higher than a reference sample.
  • sample is obtained from the subject under study.
  • sample as used herein, relates to any sample which can be obtained from the patient.
  • the present method can be applied to any type of biological sample from a patient, such as a biopsy sample, tissue, cell or fluid (serum, saliva, semen, sputum, cerebral spinal fluid (CSF), tears, mucus, sweat, milk, brain extracts and the like).
  • said sample is a tissue sample or portion thereof, preferably tumour tissue sample or portion thereof.
  • Said sample can be obtained by conventional methods, e.g., biopsy, by using methods well known to those of ordinary skill in the related medical arts.
  • Methods for obtaining the sample from the biopsy include gross apportioning of a mass, or microdissection or other art-known cell-separation methods.
  • Tumour cells can additionally be obtained from fine needle aspiration cytology. In order to simplify conservation and handling of the samples, these can be formalin-fixed and paraffin-embedded or first frozen and then embedded in a cryosolidifiable medium, such as OCT-Compound, through immersion in a highly cryogenic medium that allows for rapid freeze.
  • the first method comprises the determination of the levels of acid ceramidase.
  • the "levels of acid ceramidase” can be determined by measuring the levels of the mRNA coding acid ceramidase, by determining the levels of the acid ceramidase or by measuring the enzymatic activity of acid ceramidase.
  • the biological sample may be treated to physically or mechanically disrupt tissue or cell structure, to release intracellular components into an aqueous or organic solution to prepare nucleic acids for further analysis.
  • the nucleic acids are extracted from the sample by procedures known to the skilled person and commercially available.
  • RNA is then extracted from frozen or fresh samples by any of the methods typical in the art, for example, Sambrook, J., et al., 2001. Molecular cloning: a Laboratory Manual, 3rd ed., Cold Spring Harbor Laboratory Press, N.Y., Vol. 1-3 . Preferably, care is taken to avoid degradation of the RNA during the extraction process.
  • the expression level is determined using mRNA obtained from a formalin-fixed, paraffin-embedded tissue sample.
  • mRNA may be isolated from an archival pathological sample or biopsy sample which is first deparaffinized.
  • An exemplary deparaffinization method involves washing the paraffinized sample with an organic solvent, such as xylene.
  • Deparaffinized samples can be rehydrated with an aqueous solution of a lower alcohol. Suitable lower alcohols, for example include, methanol, ethanol, propanols, and butanols.
  • Deparaffinized samples may be rehydrated with successive washes with lower alcoholic solutions of decreasing concentration, for example. Alternatively, the sample is simultaneously deparaffinized and rehydrated. The sample is then lysed and RNA is extracted from the sample.
  • the gene mRNA expression levels are often determined by reverse transcription polymerase chain reaction (RT-PCR).
  • RT-PCR reverse transcription polymerase chain reaction
  • the expression levels of the acid ceramidase mRNA are determined by quantitative PCR, preferably, Real-Time PCR. The detection can be carried out in individual samples or in tissue microarrays.
  • Control RNA relates to a RNA whose expression levels do not change or change only in limited amounts in tumour cells with respect to non-tumourigenic cells.
  • the control RNA is mRNA corresponding to housekeeping genes and which code for proteins which are constitutively expressed and carry out essential cellular functions.
  • housekeeping genes for use in the present invention include ⁇ -2-microglobulin, ubiquitin, 18-S ribosomal protein, cyclophilin, GAPDH and actin.
  • control RNA is ⁇ -actin mRNA.
  • relative gene expression quantification is calculated according to the comparative Ct method using ⁇ -actin as an endogenous control and commercial RNA controls as calibrators. Final results, are determined according to the formula 2-( ⁇ Ct sample- ⁇ Ct calibrator), where ⁇ CT values of the calibrator and sample are determined by subtracting the CT value of the target gene from the value of the housekeeping gene.
  • the first method involves comparing the expression levels with those found in a reference sample.
  • reference sample a sample showing reference levels of acid ceramidase mRNA.
  • the reference sample maybe a tumor sample obtained from a patient similar to the tumor of the patient under study but which is not resistant to ChoK inhibitors.
  • the reference sample may be a pool of tumor tissues samples derived from several patients suffering from the same type of tumor which is under study.
  • the reference values for "increased" or “decreased” acid ceramidase mRNA levels are determined by calculating percentiles by conventional means involving the testing of a group of samples isolated from normal subj ects (i.e. people with no diagnosis of NSCLC) for the expression levels of the acid ceramidase mRNA.
  • the "increased" levels can then be assigned, preferably, to samples wherein expression levels for the acid ceramidase mRNA are equal to or in excess of percentile 50 in the normal population, including, for example, expression levels equal to or in excess to percentile 60 in the normal population, equal to or in excess to percentile 70 in the normal population, equal to or in excess to percentile 80 in the normal population, equal to or in excess to percentile 90 in the normal population, and equal to or in excess to percentile 95 in the normal population.
  • the levels of acid ceramidase can be determined by measuring the levels of the acid ceramidase protein.
  • the determination of the expression levels of the proteins can be carried out by immunological techniques such as ELISA, Western Blot or immunofluorescence.
  • Western blot is based on the detection of proteins previously resolved by gel electrophoreses under denaturing conditions and immobilized on a membrane, generally nitrocellulose by the incubation with an antibody specific and a developing system (e.g. chemoluminiscent).
  • the analysis by immunofluorescence requires the use of an antibody specific for the target protein for the analysis of the expression.
  • ELISA is based on the use of antigens or antibodies labelled with enzymes so that the conjugates formed between the target antigen and the labelled antibody results in the formation of enzymatically-active complexes. Since one of the components (the antigen or the labelled antibody) are immobilised on a support, the antibody-antigen complexes are immobilised on the support and thus, it can be detected by the addition of a substrate which is converted by the enzyme to a product which is detectable by, e.g. spectrophotometry or fluorometry.
  • the determination of the acid ceramidase protein expression levels can be carried out by constructing a tissue microarray (TMA) containing the patient samples assembled, and determining the expression levels of the protein protein by immunohistochemistry techniques.
  • Immunostaining intensity can be evaluated by two different pathologists and scored using uniform and clear cut-off criteria, in order to maintain the reproducibility of the method. Discrepancies can be resolved by simultaneous re-evaluation. Briefly, the result of immunostaining can be recorded as negative expression (0) versus positive expression, and low expression (1+) versus moderate (2+) and high (3+) expression, taking into account the expression in tumoral cells and the specific cut-off for each marker. As a general criterion, the cut-offs were selected in order to facilitate reproducibility, and when possible, to translate biological events.
  • any antibody or reagent known to bind with high affinity to the target proteins can be used for detecting the amount of target proteins. It is preferred nevertheless the use of antibody, for example polyclonal sera, hybridoma supernatants or monoclonal antibodies, antibody fragments, Fv, Fab, Fab' y F(ab')2, ScFv, diabodies, triabodies, tetrabodies and humanised antibodies.
  • a reference sample may correspond to a sample obtained by pooling equal amounts from a number of healthy patients. Once this reference sample value is established, the level of this marker expressed in tumor tissues from patients can be compared with this median value, and thus be assigned a level of "low,” "normal” or "high".
  • the collection of samples from which the reference level is derived will preferably be constituted from healthy persons from the same age as the patients. In any case it can contain a different number of samples. In a more preferred embodiment, the samples are biopsy brain samples. Preferably the collection should be sufficient to provide an accurate reference level.
  • the number of samples used for make the reference values is more than 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 500 samples.
  • an increase in expression above the reference value of at least 1.1-fold, 1.5-fold, 5-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 60-fold, 70-fold, 80-fold, 90-fold, 100-fold or even more compared with the reference value is considered as "high” expression.
  • a decrease in expression below the reference value of at least 0.9-fold, 0.75-fold, 0.2-fold, 0.1-fold, 0.05-fold, 0.025-fold, 0.02-fold, 0.01-fold, 0.005-fold or even less compared with the reference value is considered as "low" expression.
  • the determination of the protein expression levels can be carried out by constructing a tissue microarray (TMA) containing the subject samples assembled, and determining the expression levels of the proteins by immunohistochemistry techniques well known in the state of the art.
  • TMA tissue microarray
  • the determination of acid ceramidase levels is carried out by the determination of acid ceramidase activity in the sample under study.
  • Methods for the determination of the enzymatic activity of acid ceramidase are abundantly known to the skilled person and have been described in detail above.
  • a method for selecting a personalised therapy for a patient suffering from cancer comprising determining the levels of acid ceramidase in a sample from said patient wherein if the expression levels of acid ceramidase in said sample are higher than in the reference sample, the patient is candidate for being treated with a a ChoK inhibitor or a with a combination of a ChoK inhibitor and an acid ceramidase inhibitor.
  • the steps of the second method are essentially as described in the first method and includes the determination of the acid ceramidase levels in a sample from the patient (preferably a tumor sample) wherein said levels can be determined by either determining the mRNA levels, the protein levels or the acid ceramidase activity using any of the method previously described.
  • the cancer is non-small cell lung cancer.
  • third method of the invention for the identification of compounds capable of increasing the therapeutic effect of a ChoK inhibitor for the treatment of cancer comprising the steps of
  • the third method comprises a first step which consists on contacting a tumor cell showing resistance to ChoK inhibitors with a candidate compound. It will be appreciated that said contacting step can be carried out in vivo in a non-human animal which contains a tumor formed by cells resistant to ChoK inhibitors or can be carried out in vitro on a culture of cells showing resistance to ChoK inhibitors.
  • a cell culture of tumor cells resistant to ChoK inhibitors is required.
  • the cultures can be obtained from tumor cells which have been previously selected based on their resistance to ChoK inhibitors, from tumor cells previously subjected to one or more rounds of selection with increasing concentrations of a ChoK inhibitor, with cells which overexpress ChoK or with cells that constitutively express siRNA specific for ChoK. If the cells are selected by one or more rounds of selection with increasing concentrations of ChoK inhibitors, any of the ChoK inhibitors mentioned in the previous paragraphs will be adequate for this purpose.
  • a culture of ChoK-inhibitor resistant cells is established, the culture is put into contact with the candidate compound whose effect on increasing ChoK inhibitor therapeutic effect is to be measured.
  • "putting in contact" a cell with the candidate compound includes any possible way of taking the candidate compound inside the cell expressing the DNA construct.
  • the candidate compound is a molecule with low molecular weight, it is enough to add said molecule to the culture medium.
  • the candidate compound is a molecule with a high molecular weight (for example, biological polymers such as a nucleic acid or a protein), it is necessary to provide the means so that this molecule can access the cell interior.
  • the candidate molecule is a nucleic acid
  • conventional transfection means can be applied using any of the methods known in the art (calcium phosphate, DEAE-dextran, polybrene, electroporation, microinjection, liposome-mediated fusion, lipofection, infection by retrovirus and biolistic transfection).
  • the candidate compound is a protein
  • the cell can be put in contact with the protein directly or with the nucleic acid encoding it coupled to elements allowing its transcription / translation once they are in the cell interior.
  • a variant of the protein to be studied which has been modified with a peptide which can promote the translocation of the protein to the cell interior such as the Tat peptide derived from the HIV-1 TAT protein, the third helix of the Antennapedia homeodomain protein from D.melanogaster, the VP22 protein of the herpes simplex virus and arginine oligomers ( Lindgren, A. et al., 2000, Trends Pharmacol. Sci, 21:99-103 , Schwarze, S.R. et al., 2000, Trends Pharmacol. Sci., 21:45-48 , Lundberg, M et al., 2003, Mol. Therapy 8:143-150 and Snyder, E.L. and Dowdy, S.F., 2004, Pharm. Res. 21:389-393 ).
  • a variant of the protein to be studied which has been modified with a peptide which can promote the translocation of the protein to the cell interior such as the Tat peptide derived from the HIV-1 TAT
  • the compound to be assayed is preferably not isolated but forms part of a more or less complex mixture derived from a natural source or forming part of a library of compounds.
  • libraries of compounds which can be assayed according to the method of the present invention include, but are not limited to, libraries of peptides including both peptides and peptide analogs comprising D-amino acids or peptides comprising non-peptide bonds, libraries of nucleic acids including nucleic acids with phosphothioate type non-phosphodiester bonds or peptide nucleic acids, libraries of antibodies, of carbohydrates, of compounds with a low molecular weight, preferably organic molecules, of peptide mimetics and the like.
  • the library can have been preselected so that it contains compounds which can access the cell interior more easily.
  • the compounds can thus be selected based on certain parameters such as size, lipophilicity, hydrophilicity, capacity to form hydrogen bonds.
  • the compounds to be assayed can alternatively form part of an extract obtained from a natural source.
  • the natural source can be an animal, plant source obtained from any environment, including but not limited to extracts of land, air, marine organisms and the like.
  • the third method comprises the determination of the acid ceramidase levels of the cells treated with the candidate compound.
  • the determination of the acid ceramidase levels can be performed, as previously described, by determining the mRNA levels, the protein levels or the acid ceramidase activity in extracts of the cells using any of the biochemical methods previously described. Those compounds which lead to a decrease in the levels of acid ceramidase will be selected as candidate componds for increasing the response of tumor cells to ChoK inhibitors.
  • the invention additionally comprises one or several steps (iii) of fractioning said mixture and the repetition of steps (i), (ii) and (iii) of the method of the invention a variable number of times until the compound of the mixture responsible for the transcription promoting activity is isolated.
  • Methods for fractioning the compounds present in a mixture include chromatography (thin layer, gas, gel molecular exclusion, affinity chromatography) crystallization, distillation, filtration, precipitation, sublimation, extraction, evaporation, centrifugation, mass spectroscopy, adsorption and the like.
  • the screening method is carried out in vivo in an animal model of cancer obtained by implanting into a non-human animal tumor cells showing a resistance to the ChoK inhibitor.
  • the cells can be obtained using any of the methods previously mentioned.
  • the ChoK inhibitor-resistant cells can be implanted into any non-human animal of any species, preferably mammals and, more preferably, primate (monkey, baboon, chimpanzee and the like), rodent (mouse, rat, rabbit, guinea pig, hamster, and the like) or a pig.
  • the animal may be an immunodeficient animal.
  • Tumor cells that can be implanted into the recipient organism is includes circulating tumour cells, tumour stem cells, cell lines derived from the immortalisation of circulating tumour cells, micrometastatic tumour cells, cell lines derived from the immortalisation of micrometastatic tumour cells, cell lines derived from immortalized tumour cells that had been previously purified from solid tumours, primary tumour cells from solid tumours, a piece of fresh tumour that has been resected from a solid tumour, primary tumour cells, cell lines derived from immortalized cells that had been previously purified from clinical metastasis (i.e. the PC3 cell line) and any combination of any of those.
  • PC3 cell line clinical metastasis
  • the first step of the method comprising contacting said tumor cells with a candidate compound.
  • the contacting step is carried out by administering the candidate compound to the animal under conditions adequate for the compound to access the tumor cells.
  • the administration of the test compounds can be performed by any suitable route, including, for example, oral, transdermal, intravenous, infusion, intramuscular, etc. administration.
  • the expression levels of acid ceramidase in said tumor cells is determined as previously described, i.e. by determining acid ceramidase mRNA levels, acid ceramidase protein levels or acid ceramidase activity.
  • the third method of the invention involves comparing the levels of acid ceramidase in the tumor cells after treatment with the candidate compound than the levels observed prior to the treatment.
  • the acid ceramidase are lower when they show a decrease of at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 100%, i.e the acid ceramidase levels are indetectable.
  • Resected tissues from NSCLC patients were dissociated (Cell dissociation sieve-tissued grinder Kit CD1, SIGMA), and the obtained cells were seeded in 24 well plates (BD, Falcon, Bioscience, San Jose, CA, USA). Cells were treated with increasing concentrations (0, 0.5, 1, 5, 10 and 20 ⁇ M) of cDDP, Taxol, Vinolrelbine, Gemcitabine and MN58b for 10 days in DMEN:F12HAM (Ref:D8437, SIGMA) supplemented with 10% Fetal Bovine Serum (FBS, Life Technologies, Grand Island, NY). The final persistent population in each well was quantified by the method of Cristal Violet as previously described ( Rodriguez-González, A. et al., Oncogene, 22:8803-8812 ).
  • MN58b has been described in WO9805644 and corresponds to 1,4-(4-4'-Bis-((4-(dimethylamine)pyridinium-1-yl) methyl)diphenyl)butane dibromide.
  • RSM-932A has been described in US patent application US2007185170 and corresponds to 1,1'-(biphenyl-4, 4'-diylmethylene) bis [4 - (4 - chloro - N - methylanilino-) quinolinium]dibromide.
  • NOE has been described by Sugita et al (Biochim.Biphys.Acta, 1975, 398:125-131 ) and corresponds to N-oleoylethanolamine (NOE).
  • D-NMAPPD corresponds to 3-ethoxy-1-(2-methylaminoethyl)-3-phenyl-indol-2-one (CAS 35922-06-6) and has been described by Raisova, M., et al. (FEBS Lett., 2002, 516:47-52 ) and Selzner, M. et al. (Cancer Res., 2001, 61:1233-1240 ).
  • TRAIL has been described previously and corresponds to the extracellular domain of human TRAIL (amino acids 95-281).
  • RNAs from the biopsies selected were isolated for microarrays and QT-PCR, using RNeasy Mini Kit (QIAGEN, Hilden, Germany) following the instructions of the manufactures. Samples were prepared and the array hybridized according to the Affymetrix GeneChip Expression Analysis Technical Manual. Hybridization to Affymetrix U133plus2 GeneChips (54,614 probe sets, representing 47,000 transcripts), staining, washing and scanning procedures were carried out at the Genomic Facility in the National Center of Biotechnology (Madrid, Spain) as described in www.affymetrix.com (Affymetrix, Santa Clara, CA).
  • the Signal Log Ratio estimates the magnitude and direction of change of a transcript. The log scale used is base 2, thus a Signal Log Ratio of 1.0 indicates an increase of the transcript level by 2 fold and -1.0 indicates a decrease by 2 fold. A Signal Log Ratio of zero would indicate no change.
  • RNA 1 ⁇ g was used to generate cDNA using High-Capacity cDNA Archive Kit (Applied Biosystems), and quantitative real-time PCR was carried out in triplicate using the ABI PRISM 7700 Sequence Detector (Applied Biosystems). GAPDH and 18S ribosomal mRNA were amplified as internal controls.
  • Probes used for amplification were from Applied Biosystems as Taqman Gene Expression Assays (ASAH1: HS00602774_M1 Taqman Probe, ASAH2: HS00184096_M1 Taqman Probe and ASAH3: HS00370322_M1 Taqman Probe, DUT: HS00798995_S1 Taqman Probe, TYMS: HS00426591_M1 Taqman Probe, UPP1: HS00427695_M1 Taqman Probe, RRM2: HS00357247_G1 Taqman Probe).
  • the 2- ⁇ Ct method was used ( Livak KJ., Methods. 2001; 25:402-8 ) to calculate the relative expression of each gene.
  • MN58R and RSM-932A-R were generated by prolonged continuous exposure to increasing concentrations of each drug.
  • a parallel control (H460 stock) of the cell line in the absence of the compounds was kept in culture for the same time.
  • Cells were seeded on 96-well plates (BD, Falcon, Bioscience, San Jose, CA, USA) at a density of 6000 cells/well, and incubated for 24 h under standard conditions. Then, cells were treated with different concentrations of the ChoK inhibitors (quadruplicates of each concentration) and maintained for 72 hours. Quantification of the number of cells remaining in each well was carried out by the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) method. Absorbance is read at 595nm in a VersaMax Microplate Reader (Molecular Devices, Sunnyvale, CA, USA).
  • NOE N-Oleoyl ethanolamine
  • Stadistical analysis of significant changes in tumour growth was calculated using SPSS software v. 13.0.
  • Cell death was analysed by flow cytometry using de Aposcreen Annexin V Apoptosis Kit (Southern Biotech) according to manufacturer procedures. Cells were seeded at 4x10 5 cells per well in six-well. Twenty-four hours later, cells were treated with the indicated compounds. Cells were rinsed twice in cold PBS and after removing it cells were resuspended in cold 1X binding buffer to a concentration from 1x10 6 to 1x10 7 cells/mL. 100 ⁇ l of these cells were collected in a flow cytometry tube and 10 ⁇ l of Annexin V were added. After gently vortexing, tubes were incubated for 15 minutes on ice, protected from light. Without washing, 380 ⁇ l of 1X binding buffer were added to each sample prior to 10 ⁇ l of propidium iodine. Cells were immediately analysed by flow cytometry.
  • lipids were seeded at 1x10 5 cells per well in six-well plates and lipids were labelled with [ 14 C]-Serine for two days. Cells were treated without replacing the labelled medium for 24 hours. Then lipids were extracted and analysed by TLC using a previously described protocol ( van Echten-Deckert, G. (2000) Sphingolipid extraction and analysis by thin-layer chromatography. Methods Enzymol, 312, 64-79 ). Prior to lipid extraction, cells were kept at -20 °C. Cells were harvested in methanol and lipids were extracted in a final ratio of chloroform-methanol-water of 60:30:6 (v/v/v). The organic phase was dried in a concentrator under N 2 flow at 37 °C.
  • the liquid chromatography-mass spectrometer consisted of a Waters Aquity UPLC system connected to a Waters LCT Premier orthogonal accelerated time of flight mass spectrometer (Waters, Millford, MA), operated in positive electrospray ionisation mode. Full scan spectra from 50 to 1500 Da were acquired and individual spectra were summed to produce data points each 0.2 s. Mass accuracy and reproducibility were maintained by using an independent reference spray via the LockSpray interference.
  • the analytical column was a 100 mm ⁇ 2.1mm id, 1.7 ⁇ m C8 Acquity UPLC BEH (Waters).
  • phase A MeOH/H 2 O/HCOOH (74:25:1 v/v/v); phase B: MeOH/HCOOH (99/1 v/v), both also contained 5 mM ammonium formate.
  • a gradient was programmed-0.0 min, 80% B; 3 min, 90% B; 6 min, 90% B; 15 min, 99% B; 18 min, 99% B; 20 min, 80% B.
  • the flow rate was 0.3 mL min -1 .
  • the column was held at 30 °C. Quantification was carried out using the extracted ion chromatogram of each compound, using 50 mDa windows. The linear dynamic range was determined by injecting standard mixtures. Positive identification of compounds was based on the accurate mass measurement with an error ⁇ 5 ppm and its LC retention time, compared to that of a standard ( ⁇ 2%).
  • MTT growth assays as described above were used to evaluate cisplatin in combination with ChoK inhibitors. Following an overnight incubation, cisplatin was added in varying concentrations and incubated for 3h. ChoK inhibitors in growing concentrations were then added for 40h, and afterwards, cells were cultured for additional 24 hours in fresh culture medium.
  • the results from combination assays in terms of synergy, additivity or antagonism were analyzed using the isobologram combination index method of Chou Talalay ( Chou TC. et al., Trends Pharmacol Sci, 1983, 4:450-4 ). The ranges of CI are established from those already described (Chou TC. et al., supra .).
  • acid ceramidase an enzyme involved in the lipid metabolism was found significantly over-expressed according to any selection criteria in those tumours that were resistant to MN58b.
  • This latter metabolite is a potent inducer of cell death.
  • Resistance to the action of ChoKI can be produced if the activity of the enzyme responsible for the cnversion of ceramides into esphingosine is increased.
  • This enzyme is acid ceramidase (ASAH1) ( Figure 3 ).
  • CAY10466 (CAY) (D-NMAPPD (1R,2R)-B13; 10006305 Cayman Chemical) (item VIII in Table 2) and an alkaline ceramidase inhibitor as a negative control D-erythro-MAPP (DMP) (10165 Cayman Chemical).
  • DMP D-erythro-MAPP
  • Choline kinase inhibitors and TRAIL cooperates to induce cell death in colon cancer derived cell lines.
  • cells were seeded in 96 multiwell plates at a density of 1x10 4 cells/well 24 hours before treatment.
  • Cells were treated with different concentrations of RSM-932A (ChoKI) or TRAIL for different times and the cell proliferation was quantified using the MTT [3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide] colorimetric assay.
  • TRAIL is a member of the tumour necrosis factor (TNF) superfamily and can induce apoptosis by the extrinsic pathway associated via stimulation of death receptors DR4 (TNFRSF10A or TRAIL-R1) and DR5 (TNFRSF10B or TRAIL-R2).
  • DR4 TNFRSF10A or TRAIL-R1
  • DR5 TNFRSF10B or TRAIL-R2
  • DR5 and DR4 levels were analysed by quantitative PCR in DLD-1 and SW620 cell lines after treatments with MN58b, TRAIL or their simultaneous combination.
  • Choline kinase inhibitors and 5-FU cooperates to induce cell death in colon cancer derived cell lines.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Analytical Chemistry (AREA)
  • Epidemiology (AREA)
  • Wood Science & Technology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • Physics & Mathematics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Hematology (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • Pathology (AREA)
  • Genetics & Genomics (AREA)
  • Oncology (AREA)
  • Hospice & Palliative Care (AREA)
  • General Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • General Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Cell Biology (AREA)
  • Inorganic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Gastroenterology & Hepatology (AREA)
EP09783138.2A 2008-09-17 2009-09-17 Methods and compositions for the treatment of cancer Active EP2340042B1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
PL09783138T PL2340042T3 (pl) 2008-09-17 2009-09-17 Sposoby i kompozycje do leczenia raka
EP09783138.2A EP2340042B1 (en) 2008-09-17 2009-09-17 Methods and compositions for the treatment of cancer

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP08380270 2008-09-17
PCT/EP2009/062078 WO2010031825A2 (en) 2008-09-17 2009-09-17 Methods and compositions for the treatment of cancer
EP09783138.2A EP2340042B1 (en) 2008-09-17 2009-09-17 Methods and compositions for the treatment of cancer

Publications (2)

Publication Number Publication Date
EP2340042A2 EP2340042A2 (en) 2011-07-06
EP2340042B1 true EP2340042B1 (en) 2014-07-23

Family

ID=40364454

Family Applications (1)

Application Number Title Priority Date Filing Date
EP09783138.2A Active EP2340042B1 (en) 2008-09-17 2009-09-17 Methods and compositions for the treatment of cancer

Country Status (16)

Country Link
US (4) US20100068302A1 (pt)
EP (1) EP2340042B1 (pt)
JP (1) JP2012502954A (pt)
KR (1) KR20110067041A (pt)
CN (1) CN102215872B (pt)
AU (1) AU2009294618A1 (pt)
BR (1) BRPI0919238A2 (pt)
CA (1) CA2737551A1 (pt)
DK (1) DK2340042T3 (pt)
ES (1) ES2527361T3 (pt)
HK (1) HK1159498A1 (pt)
MX (1) MX2011002956A (pt)
PL (1) PL2340042T3 (pt)
PT (1) PT2340042E (pt)
RU (1) RU2011143520A (pt)
WO (1) WO2010031825A2 (pt)

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8889127B2 (en) 2004-07-01 2014-11-18 Icahn School Of Medicine At Mount Sinai Targeted protein replacement for the treatment of lysosomal storage disorders
WO2012051415A2 (en) * 2010-10-13 2012-04-19 Mount Sinai School Of Medicine Inhibitors of acid ceramidase and uses thereof in cancer and other disease treatment therapies
EP2468259A1 (en) * 2010-12-23 2012-06-27 Traslational Cancer Drugs Pharma, S.L. Pharmaceutical compositions of pyridinium and quinolinium derivatives
EP2721174A1 (en) 2011-06-20 2014-04-23 Traslational Cancer Drugs Pharma, S.L. Method for predicting the clinical response to chemotherapy in a subject with cancer
DK2753346T3 (da) 2011-09-07 2020-05-25 Sinai School Medicine Ceramidase og celledifferentiering
EP3679923B1 (en) * 2012-06-01 2021-08-04 Icahn School of Medicine at Mount Sinai Ceramide levels in the treatment and prevention of infections
US20150182490A1 (en) 2012-06-26 2015-07-02 Del Mar Pharmaceuticals Methods for treating tyrosine-kinase-inhibitor-resistant malignancies in patients with genetic polymorphisms or ahi1 dysregulations or mutations employing dianhydrogalactitol, diacetyldianhydrogalactitol, dibromodulcitol, or analogs or derivatives thereof
US11813338B2 (en) 2013-03-12 2023-11-14 The Trustees Of The University Of Pennsylvania Diagnosing and treating cancer
JP6832158B2 (ja) 2013-03-14 2021-02-24 アイカーン スクール オブ メディシン アット マウント サイナイ 治療用酸性セラミダーゼ組成物ならびにそれを作製および使用する方法
JP2016519684A (ja) 2013-04-08 2016-07-07 デニス エム ブラウン 準最適に投与された薬物療法の有効性を改善するための及び/又は副作用を低減するための方法および組成物
KR101537030B1 (ko) * 2013-08-23 2015-07-16 (주)바이오디펜스 천적동물의 눈을 포함한 홀로그램을 이용한 농작물 보호방법 및 그 보호기구
WO2015028662A1 (en) 2013-08-30 2015-03-05 Consejo Superior De Investigaciones Cientificas (Csic) Compositions and methods for characterization and amelioration of rheumatoid arthritis
US11299528B2 (en) 2014-03-11 2022-04-12 D&D Pharmatech Inc. Long acting TRAIL receptor agonists for treatment of autoimmune diseases
CA3008392C (en) 2015-12-17 2021-11-09 The Johns Hopkins University Ameliorating systemic sclerosis with death receptor agonists
EA201892260A1 (ru) 2016-04-07 2019-03-29 Дзе Джонс Хопкинс Юниверсити Композиции и способы для лечения панкреатита и боли с применением агонистов рецептора смерти
CN107362370B (zh) * 2016-05-13 2022-07-26 国家纳米科学中心 一种基于金纳米簇联合NGF siRNA治疗胰腺癌的方法
CN109744199A (zh) * 2017-11-08 2019-05-14 南京艾莫瑞生物科技有限公司 一种肿瘤细胞异种移植斑马鱼模型、其构建方法及应用
WO2021035168A1 (en) * 2019-08-22 2021-02-25 Thomas Jefferson University Methods for reprogramming cancer cells
CN114644626A (zh) * 2020-12-18 2022-06-21 南京施江医药科技有限公司 一种苯环类化合物及其应用

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5830916A (en) * 1996-05-23 1998-11-03 Duke University Inhibitor of ceramidase
ES2117950B1 (es) * 1996-08-02 1999-09-16 Univ Granada Nuevos compuestos que bloquean la biosintesis de fosforilcolina y su uso como segundo mensajero en proliferacion celular.
US6511676B1 (en) * 1999-11-05 2003-01-28 Teni Boulikas Therapy for human cancers using cisplatin and other drugs or genes encapsulated into liposomes
GB0121285D0 (en) * 2001-09-03 2001-10-24 Cancer Res Ventures Ltd Anti-cancer combinations
ES2237332B1 (es) * 2004-01-14 2006-11-01 Consejo Sup. Investig. Cientificas Derivados de piridinio y quinolinio.
AU2005249206A1 (en) * 2004-06-03 2005-12-15 F. Hoffmann-La Roche Ag Treatment with cisplatin and an EGFR-inhibitor
PT1889920E (pt) * 2005-04-13 2010-04-14 Consejo Superior Investigacion Método para a identificação in vitro de compostos para terapia do cancro
ES2277568B1 (es) * 2005-12-30 2008-04-01 Consejo Superior De Investigaciones Cientificas Derivados de triterpenoquinona y triterpenofenoles y su aplicacion para el tratamiento de tumores y enfermedades parasitarias.

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
K. GLUNDE: "RNA Interference-Mediated Choline Kinase Suppression in Breast Cancer Cells Induces Differentiation and Reduces Proliferation", CANCER RESEARCH, vol. 65, no. 23, 1 December 2005 (2005-12-01), pages 11034 - 11043, XP055067870, ISSN: 0008-5472, DOI: 10.1158/0008-5472.CAN-05-1807 *
RAMIREZ DE MOLINA A ET AL: "Choline kinase as a link connecting phospholipid metabolism and cell cycle regulation: Implications in cancer therapy", INTERNATIONAL JOURNAL OF BIOCHEMISTRY AND CELL BIOLOGY, PERGAMON, GB, vol. 40, no. 9, 1 January 2008 (2008-01-01), pages 1753 - 1763, XP022734731, ISSN: 1357-2725, [retrieved on 20080119], DOI: 10.1016/J.BIOCEL.2008.01.013 *

Also Published As

Publication number Publication date
WO2010031825A3 (en) 2010-08-19
US20120040021A1 (en) 2012-02-16
RU2011143520A (ru) 2013-05-10
CN102215872A (zh) 2011-10-12
PL2340042T3 (pl) 2015-01-30
AU2009294618A2 (en) 2011-04-14
CN102215872B (zh) 2015-06-10
US20110256241A1 (en) 2011-10-20
DK2340042T3 (da) 2014-10-27
HK1159498A1 (en) 2012-08-03
AU2009294618A1 (en) 2010-03-25
US20100068302A1 (en) 2010-03-18
KR20110067041A (ko) 2011-06-20
EP2340042A2 (en) 2011-07-06
MX2011002956A (es) 2011-08-03
US20150297576A1 (en) 2015-10-22
JP2012502954A (ja) 2012-02-02
WO2010031825A2 (en) 2010-03-25
ES2527361T3 (es) 2015-01-22
BRPI0919238A2 (pt) 2019-09-24
CA2737551A1 (en) 2010-03-25
PT2340042E (pt) 2014-11-27

Similar Documents

Publication Publication Date Title
EP2340042B1 (en) Methods and compositions for the treatment of cancer
Parsels et al. Gemcitabine sensitization by checkpoint kinase 1 inhibition correlates with inhibition of a Rad51 DNA damage response in pancreatic cancer cells
Lee et al. The immunohistochemical overexpression of ribonucleotide reductase regulatory subunit M1 (RRM1) protein is a predictor of shorter survival to gemcitabine-based chemotherapy in advanced non-small cell lung cancer (NSCLC)
Gandhirajan et al. Small molecule inhibitors of Wnt/β-catenin/lef-1 signaling induces apoptosis in chronic lymphocytic leukemia cells in vitro and in vivo
US20040167139A1 (en) Methods of treating cancer
JP2019531699A (ja) Nrf2及びその遺伝子の下流標的遺伝子の発現状態及び変異状態によるがんの診断及び治療方法
CN111373055A (zh) 用于癌症的诊断和治疗方法
JP2005529616A (ja) 化学療法耐性腫瘍の診断および治療
EP1845094A1 (en) Method of tumor suppression and evaluation of anticancer agent based on gsk3 beta inhibitory effect
Huang et al. Characterization of the biological activity of a potent small molecule Hec1 inhibitor TAI-1
Yuan et al. TIPE3 is a regulator of cell apoptosis in glioblastoma
Elshaer et al. Leflunomide-induced liver injury in mice: involvement of TLR4 mediated activation of PI3K/mTOR/NFκB pathway
US20090163564A1 (en) Translational Dysfunction Based Therapeutics
WO2012175537A1 (en) Method for predicting the clinical response to chemotherapy in a subject with cancer
EP1971690B1 (en) Translational dysfunction based therapeutics
EP2979708A1 (en) Composition for treatment or metastasis suppression of cancers which includes p34 expression inhibitor or activity inhibitor as active ingredient
EP2082235B1 (en) A method of diagnosis and agents useful for same
JP6975720B2 (ja) 腫瘍の診断および治療におけるAkt2の使用
Mukherjee et al. Ca2+/Calmodulin Dependent Protein Kinase Kinase-2 (CaMKK2) promotes Protein Kinase G (PKG)-dependent actin cytoskeletal assembly to increase tumor metastasis
JP2015511598A (ja) Pak1阻害剤を用いて黒色腫を治療する方法
US20140363449A1 (en) Methods, assays, and systems relating to sakt
US20120121610A1 (en) Therapeutic agent and assay
JP2008230977A (ja) ニチジンを成分とする抗癌剤、および該抗癌剤の感受性増強剤
JP2022028749A (ja) 腫瘍の診断および治療におけるAkt2の使用

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20110414

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK SM TR

AX Request for extension of the european patent

Extension state: AL BA RS

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1159498

Country of ref document: HK

17Q First examination report despatched

Effective date: 20121030

RIC1 Information provided on ipc code assigned before grant

Ipc: A61K 45/06 20060101AFI20131203BHEP

Ipc: A61K 31/4425 20060101ALI20131203BHEP

Ipc: A61P 35/00 20060101ALI20131203BHEP

Ipc: A61K 33/24 20060101ALI20131203BHEP

Ipc: C12Q 1/34 20060101ALI20131203BHEP

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20140211

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: TRASLATIONAL CANCER DRUGS PHARMA, S.L.

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK SM TR

AX Request for extension of the european patent

Extension state: AL BA RS

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 678493

Country of ref document: AT

Kind code of ref document: T

Effective date: 20140815

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602009025533

Country of ref document: DE

Effective date: 20140904

REG Reference to a national code

Ref country code: RO

Ref legal event code: EPE

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

Effective date: 20141021

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: NL

Ref legal event code: T3

REG Reference to a national code

Ref country code: PT

Ref legal event code: SC4A

Free format text: AVAILABILITY OF NATIONAL TRANSLATION

Effective date: 20141117

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

REG Reference to a national code

Ref country code: GR

Ref legal event code: EP

Ref document number: 20140402153

Country of ref document: GR

Effective date: 20141121

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2527361

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20150122

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141023

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141023

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140723

REG Reference to a national code

Ref country code: PL

Ref legal event code: T3

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140723

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140723

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20141123

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140723

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602009025533

Country of ref document: DE

REG Reference to a national code

Ref country code: HU

Ref legal event code: AG4A

Ref document number: E022311

Country of ref document: HU

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140723

Ref country code: LU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140917

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140723

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140723

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20141023

26N No opposition filed

Effective date: 20150424

REG Reference to a national code

Ref country code: HK

Ref legal event code: GR

Ref document number: 1159498

Country of ref document: HK

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140930

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140930

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20141023

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140723

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140723

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140723

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FI

Payment date: 20160927

Year of fee payment: 8

Ref country code: IE

Payment date: 20160927

Year of fee payment: 8

Ref country code: DK

Payment date: 20160927

Year of fee payment: 8

Ref country code: NL

Payment date: 20160926

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: HU

Payment date: 20160929

Year of fee payment: 8

Ref country code: SE

Payment date: 20160928

Year of fee payment: 8

Ref country code: RO

Payment date: 20160922

Year of fee payment: 8

Ref country code: CZ

Payment date: 20160921

Year of fee payment: 8

Ref country code: FR

Payment date: 20160926

Year of fee payment: 8

Ref country code: PT

Payment date: 20160923

Year of fee payment: 8

Ref country code: GR

Payment date: 20160928

Year of fee payment: 8

Ref country code: PL

Payment date: 20160920

Year of fee payment: 8

Ref country code: AT

Payment date: 20160921

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: TR

Payment date: 20160921

Year of fee payment: 8

Ref country code: BE

Payment date: 20160927

Year of fee payment: 8

Ref country code: ES

Payment date: 20160926

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DE

Payment date: 20160928

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IT

Payment date: 20160923

Year of fee payment: 8

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 602009025533

Country of ref document: DE

REG Reference to a national code

Ref country code: DK

Ref legal event code: EBP

Effective date: 20170930

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170917

Ref country code: CZ

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170917

Ref country code: RO

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170917

REG Reference to a national code

Ref country code: SE

Ref legal event code: EUG

REG Reference to a national code

Ref country code: NL

Ref legal event code: MM

Effective date: 20171001

REG Reference to a national code

Ref country code: AT

Ref legal event code: MM01

Ref document number: 678493

Country of ref document: AT

Kind code of ref document: T

Effective date: 20170917

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180319

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

REG Reference to a national code

Ref country code: BE

Ref legal event code: FP

Effective date: 20141022

Ref country code: BE

Ref legal event code: MM

Effective date: 20170930

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20171001

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20140723

REG Reference to a national code

Ref country code: FR

Ref legal event code: ST

Effective date: 20180531

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180404

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170917

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170918

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170930

Ref country code: AT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170917

Ref country code: IT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170917

Ref country code: GR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180403

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20171002

REG Reference to a national code

Ref country code: ES

Ref legal event code: FD2A

Effective date: 20181019

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170930

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: ES

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170918

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PL

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170917

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170918

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170917